VEGF Rat (120a.a.), Yeast

Vascular Endothelial Growth Factor (120a.a.) Rat Recombinant, Yeast
Shipped with Ice Packs
In Stock

Description

Key Biological Activities

VEGF Rat (120a.a.) promotes:

  • Angiogenesis: Stimulation of endothelial cell proliferation and migration .

  • Vasculogenesis: Induction of vascular network formation .

  • Vascular Permeability: Originally identified as a vascular permeability factor .

  • Anti-Apoptotic Effects: Inhibition of endothelial cell apoptosis .

ED50 in Functional Assays:

Assay TypeED50 RangeCell LineSource
HUVEC Proliferation2–10 ng/mLHuman Umbilical Vein Endothelial Cells

ELISA Compatibility

Rat VEGF (120a.a.) shows limited cross-reactivity with other species in immunoassays:

SpeciesCross-Reactivity (%)Source
Mouse VEGF73% (E. coli-expressed)
Human VEGF24% (E. coli-expressed)
Canine VEGF14% (Sf21-expressed)

This specificity is critical for distinguishing rat VEGF in mixed-species studies .

Therapeutic Angiogenesis

VEGF Rat (120a.a.) is used in preclinical models for ischemic diseases. A study demonstrated controlled release from alginate microparticles using ionic crosslinkers (e.g., Ca²⁺, Zn²⁺), enabling sustained bioactive delivery .

CrosslinkerRelease Profile (VEGF)Bioactivity RetentionSource
Ca²⁺Fast releaseHigh (HUVEC sprouting)
Zn²⁺Sustained releaseHigh

Role in Disease Models

  • Diabetic Retinopathy: Associated with VEGF mutations and pathological angiogenesis .

  • POEMS Syndrome (Crow-Fukase): Elevated VEGF levels correlate with disease progression .

Amino Acid Sequence

The sequence includes conserved regions critical for receptor binding:
MAPTTEGEQK AHEVVKFMDV YQRSYCRPIE TLVDIFQEYP DEIEYIFKPS CVPLMRCAGC CNDEALECVP TSESNVTMQI MRIKPHQSQH IGEMSFLQHS RCECRPKKDR TKPEKCDKPR R .

Disulfide Bonding

The homodimer structure is stabilized by eight conserved cysteine residues, forming a cystine knot motif characteristic of VEGF family proteins .

Product Specs

Introduction

Vascular endothelial growth factor (VEGF) is a key signaling protein that plays a vital role in angiogenesis (formation of new blood vessels) and vasculogenesis (development of the circulatory system). VEGF primarily affects vascular endothelial cells but also impacts other cell types, including monocytes/macrophages, neurons, cancer cells, and kidney epithelial cells. It contributes to increased vascular permeability, promotes angiogenesis and vasculogenesis, stimulates endothelial cell growth and migration, and inhibits apoptosis (programmed cell death). In vitro studies have demonstrated VEGF's ability to enhance endothelial cell division (mitogenesis) and migration. VEGF also acts as a vasodilator, widening blood vessels, and increases microvascular permeability, initially known as vascular permeability factor. Notably, elevated VEGF levels are associated with POEMS syndrome (Crow-Fukase syndrome). Moreover, mutations in the VEGF gene have been linked to both proliferative and nonproliferative diabetic retinopathy.

Description

Recombinant Rat Vascular Endothelial Growth Factor (120a.a.), produced in yeast, is a homodimer protein consisting of two identical polypeptide chains, each comprising 121 amino acids. The two chains are linked together by disulfide bonds, resulting in a total molecular weight of approximately 18.5 kDa per chain. The purification process involves proprietary chromatographic techniques to ensure high purity.

Physical Appearance

Sterile Filtered White lyophilized (freeze-dried) powder.

Formulation

The product is lyophilized (freeze-dried) from a concentrated solution in phosphate-buffered saline (PBS) at a pH of 7.4. The solution is sterile-filtered through a 0.2-micrometer filter before lyophilization.

Solubility

To reconstitute the lyophilized Vascular Endothelial Growth Factor, it is recommended to dissolve it in sterile 18 megaohm-centimeter (MΩ·cm) H₂O (water) at a concentration of at least 100 micrograms per milliliter (µg/ml). The reconstituted solution can then be further diluted to desired concentrations in other aqueous solutions as needed.

Stability

Lyophilized VEGF, while stable at room temperature for up to 3 weeks, should ideally be stored in a dry environment (desiccated) at a temperature below -18°C for long-term storage. After reconstitution, Vascular Endothelial Growth Factor should be stored at 4°C and used within 2-7 days. For long-term storage of the reconstituted solution, it is recommended to store it below -18°C. Avoid repeated freeze-thaw cycles to maintain protein integrity and activity.

Purity

The purity of the Vascular Endothelial Growth Factor is greater than 90%, as determined by two analytical methods:
(a) Reverse-Phase High-Performance Liquid Chromatography (RP-HPLC)
(b) Sodium Dodecyl Sulfate-Polyacrylamide Gel Electrophoresis (SDS-PAGE).

Biological Activity

The biological activity of Vascular Endothelial Growth Factor is determined by its effective dose 50 (ED₅₀), which refers to the concentration required to achieve 50% of the maximum biological response. In this case, the ED₅₀ was measured using a cell proliferation assay with HUVEC (human umbilical vein endothelial cells) and was found to be in the range of 2-10 nanograms per milliliter (ng/ml).

Synonyms

Vascular endothelial growth factor A, VEGF-A, Vascular permeability factor, VPF, VEGF, MGC70609.

Source

Saccharomyces cerevisiae

Amino Acid Sequence

MAPTTEGEQK AHEVVKFMDV YQRSYCRPIE TLVDIFQEYP DEIEYIFKPS CVPLMRCAGC CNDEALECVP TSESNVTMQI MRIKPHQSQH IGEMSFLQHS RCECRPKKDR TKPEKCDKPR R.

Q&A

What is VEGF Rat (120a.a.), Yeast and what are its primary biological functions?

VEGF Rat (120a.a.), Yeast refers to rat Vascular Endothelial Growth Factor with 120 amino acids produced in Saccharomyces cerevisiae expression systems. It is a disulfide-linked homodimer consisting of two 121 amino acid polypeptide chains, each with a molecular mass of approximately 18.5kDa. This signaling protein is primarily involved in both angiogenesis (formation of new blood vessels from pre-existing vessels) and vasculogenesis (de novo formation of blood vessels). VEGF mediates increased vascular permeability, induces endothelial cell growth, promotes cell migration, and inhibits apoptosis. It also functions as a vasodilator and increases microvascular permeability, which explains why it was originally referred to as vascular permeability factor . Though VEGF's activity has been predominantly studied in vascular endothelial cells, it also affects numerous other cell types, including monocytes/macrophages, neurons, cancer cells, and kidney epithelial cells .

How can I optimize VEGF-induced signaling studies in vitro using rat VEGF (120a.a.)?

To optimize VEGF-induced signaling studies, several methodological considerations are essential. First, cell selection is critical—while HUVEC cells are commonly used with an effective dose range of 2-10 ng/ml for rat VEGF (120a.a.) , other endothelial cell types may require different concentrations. Serum starvation (0.1% FBS in endothelial basic medium for 24 hours) prior to VEGF treatment improves signal-to-noise ratio by reducing baseline phosphorylation of signaling proteins . For western blot analysis of VEGFR2 activation and downstream molecules (ERK1/2, PLCγ), collect lysates at multiple time points (5, 15, 30, 60 minutes) after VEGF treatment to capture the full signaling dynamics . Include appropriate controls such as heat-inactivated VEGF, vehicle-only treatments, and VEGFR2-specific inhibitors (e.g., Ki-8751) . For detection of phosphorylated signaling molecules, use 4-20% gradient SDS-PAGE to achieve optimal separation . When selecting antibodies, ensure they specifically recognize rat VEGF or cross-react appropriately with your experimental system . Quantify results by normalizing phosphorylated protein signals to their respective total protein levels to account for loading variations. Finally, validate key findings using multiple detection methods such as ELISA or phospho-flow cytometry to confirm western blot results.

What are the sex-specific differences in VEGF/VEGFR2 signaling responses, and how should these be addressed in experimental design?

Sex-specific differences in VEGF/VEGFR2 signaling responses represent an important but often overlooked aspect of experimental design. Research has demonstrated distinct responses to VEGFR2 blockade between male and female rats, particularly in inflammatory conditions. In studies of chronic cyclophosphamide-induced cystitis, VEGFR2 blockade increased void volume 1.4-fold in female rats (P ≤ 0.05) but 2.2-fold in male rats (P ≤ 0.01), suggesting stronger VEGFR2-mediated regulation in males under these conditions . In control conditions, the response magnitude is more similar between sexes, with void volume increases of 1.2-fold in females and 1.3-fold in males following VEGFR2 antagonism . These findings highlight that the impact of VEGF/VEGFR2 signaling may have sex-dependent patterns that vary based on physiological context. To address these differences in experimental design, researchers should: (1) include both sexes in VEGF studies rather than assuming transferability of results, (2) power studies sufficiently to detect potential sex-specific effects, (3) analyze data disaggregated by sex before pooling, (4) consider hormonal status in female animals as estrogen can regulate VEGF expression, and (5) design dose-response studies that may reveal sex-dependent differences in sensitivity to VEGF or VEGFR2 antagonists. The growing evidence for sex-specific effects emphasizes that comprehensive VEGF research requires attention to sex as a biological variable.

How does VEGF expression and signaling change during different stages of inflammation, and what implications does this have for therapeutic targeting?

VEGF expression and signaling undergo dynamic changes during the progression of inflammation, with distinct patterns between acute and chronic phases. In models of cyclophosphamide-induced cystitis, researchers have observed tissue-specific distribution of VEGF-A isoforms (both pro-angiogenic VEGF-Axxxa and anti-angiogenic VEGF-Axxxb) that change depending on the inflammatory stage . During acute inflammation (4 hours post-induction), VEGF expression rapidly increases as part of the initial vascular response, promoting increased permeability and immune cell recruitment. In chronic inflammation (7-8 days), VEGF signaling patterns shift toward tissue remodeling and neovascularization to support the inflamed tissue . The functional consequences of these temporal changes are reflected in the differential responses to VEGFR2 blockade, where void volume increases after VEGFR2 antagonist administration in female rats with both acute and chronic cystitis, but male rats show significant responses only in the chronic condition . This suggests that the therapeutic window for VEGF/VEGFR2-targeted interventions may be inflammation stage-dependent and sex-specific. For therapeutic targeting, these observations imply that: (1) timing of anti-VEGF interventions is critical, with potentially different optimal windows for acute versus chronic inflammation; (2) biomarker assessment of VEGF isoform ratios may help predict treatment responsiveness; (3) combination therapies targeting multiple aspects of inflammation may be more effective than VEGF inhibition alone; and (4) sex-specific dosing or treatment protocols may be necessary to achieve optimal therapeutic outcomes.

What are the optimal storage and handling conditions for rat VEGF (120a.a.) to maintain its biological activity?

Proper storage and handling of rat VEGF (120a.a.) are crucial for maintaining its biological activity. The lyophilized protein should be stored at -20°C for long-term stability . Once reconstituted, the protein can be stored at 4°C for short-term use (1-2 weeks) , but for extended stability, aliquoting and storing at -80°C is recommended to prevent repeated freeze-thaw cycles. When reconstituting lyophilized VEGF, use sterile techniques and add buffer solutions gently, allowing the protein to dissolve completely without excessive agitation which can cause denaturation. Working solutions should be prepared in buffers containing carrier proteins like 0.1-0.5% BSA to minimize adsorption to plasticware and pipette tips. For cell culture applications, prepare fresh dilutions from stock solutions to ensure accurate dosing, particularly important given the potent biological activity at low concentrations (ED50 of 2-10 ng/ml in HUVEC proliferation assays) . Visual inspection should reveal a clear solution without particulate matter after reconstitution. The biological activity of VEGF preparations can be verified using established bioassays such as HUVEC proliferation or migration assays prior to use in critical experiments. If prolonged storage is necessary, stability can be enhanced by adding stabilizers like trehalose or glycerol at appropriate concentrations.

What are the recommended methods for quantifying angiogenic responses to rat VEGF (120a.a.) in in vivo models?

Quantifying angiogenic responses to rat VEGF (120a.a.) in vivo requires robust methodological approaches to capture both structural and functional vascular changes. A validated approach involves fluorescent labeling of blood vessels using intravascular injection of fluorescent markers such as Alexa Fluor® 647 WGA (50 μg) via retro-orbital administration . After a brief circulation period (approximately 7 minutes), tissues of interest should be harvested, rinsed in PBS, and immediately imaged using confocal microscopy to preserve the in vivo vascular architecture . Three-dimensional image acquisition followed by reconstruction into two-dimensional grayscale images and binarization to black-and-white vascular patterns allows for quantitative assessment of vessel density, branching patterns, and volume . This technique can be supplemented with complementary methods including: (1) quantitative PCR analysis of angiogenesis-related genes in the target tissue to correlate structural changes with molecular signatures; (2) immunohistochemical staining for endothelial markers (CD31/PECAM-1) and proliferation markers (Ki-67) to assess active angiogenesis; (3) functional perfusion assessments using laser Doppler imaging or contrast-enhanced ultrasound; and (4) vessel permeability assays using Evans blue dye extravasation or fluorescent dextran leakage. For long-term studies, implantable window chambers may allow for longitudinal tracking of angiogenic responses in the same animal. Statistical analysis should account for regional heterogeneity within tissues by analyzing multiple fields per sample and calculating both mean values and measures of spatial variation.

How can I effectively design experiments to study the cross-talk between VEGF signaling and other growth factor pathways?

Designing experiments to study cross-talk between VEGF signaling and other growth factor pathways requires strategic approaches to delineate specific interactions while maintaining physiological relevance. A comprehensive experimental design should incorporate the following elements: First, implement a sequential stimulation protocol where cells are pre-treated with one growth factor (e.g., FGF2, HGF, or IGF-1) before VEGF stimulation, with careful timing to capture both early (minutes) and sustained (hours) signaling events . Include simultaneous stimulation conditions to identify synergistic or antagonistic effects. Use pathway-specific inhibitors at multiple nodes of each signaling cascade to distinguish between direct cross-talk versus downstream convergence of separate pathways. For in vivo studies, consider using models where expression of multiple growth factors is altered, such as the metanephros transplantation model which shows increased expression of VEGF, FGF2, HGF, and IGF-1 . Employ phospho-proteomics approaches to identify novel points of intersection between pathways without bias from predetermined targets. For mechanistic validation, utilize genetic approaches (siRNA, CRISPR-Cas9) to silence specific pathway components and observe effects on cross-pathway activation. Complement protein-level analyses with transcriptomic assessments to identify gene expression signatures associated with pathway cross-talk. Additionally, consider co-immunoprecipitation studies to detect physical interactions between components of different signaling pathways. When analyzing results, develop computational models that can integrate multi-pathway data to predict emergent properties of the signaling network. This structured approach allows for comprehensive mapping of VEGF's interactions with other growth factor pathways and provides deeper insight into the complexities of angiogenic regulation.

How can rat VEGF (120a.a.) be used to develop in vitro angiogenesis assays?

Rat VEGF (120a.a.) serves as an excellent tool for developing reproducible in vitro angiogenesis assays that model distinct aspects of the angiogenic process. To establish a comprehensive angiogenesis assessment platform, researchers should implement multiple complementary assays. For endothelial cell proliferation assays, HUVECs cultured in minimal media (0.1% serum) respond to rat VEGF (120a.a.) in a dose-dependent manner, with optimal concentrations in the 2-10 ng/ml range . Cell proliferation can be quantified using BrdU incorporation, MTT/MTS assays, or automated cell counting systems. For migration assays, the scratch wound healing method offers a straightforward approach—create a consistent "wound" in a confluent HUVEC monolayer, add rat VEGF (120a.a.), and monitor closure over 8-24 hours using time-lapse microscopy. More sophisticated Boyden chamber or Transwell assays allow for chemotactic gradient assessment, with VEGF (5-50 ng/ml) in the lower chamber. Tube formation assays on Matrigel or other extracellular matrix substitutes provide a three-dimensional assessment of endothelial cell organization, with rat VEGF (120a.a.) accelerating and enhancing network formation typically within 4-16 hours of treatment. For more complex models, consider co-culture systems with supporting cells (pericytes, fibroblasts) to better recapitulate the cellular interactions in angiogenesis. The spheroid sprouting assay, where HUVEC spheroids embedded in collagen gels develop sprouts in response to VEGF, offers a quantifiable model of sprouting angiogenesis. In all these assays, parallel experiments with VEGF receptor inhibitors (such as Ki-8751) serve as important controls to confirm the specificity of the observed responses .

How does rat VEGF (120a.a.) from yeast compare to other isoforms in therapeutic angiogenesis studies?

Rat VEGF (120a.a.) from yeast presents distinct advantages and considerations in therapeutic angiogenesis studies compared to other isoforms. The 120-amino acid variant lacks the heparin-binding domains found in larger isoforms (such as VEGF164), resulting in greater diffusibility, which allows for a wider distribution from the administration site . This property makes VEGF120 particularly suitable for conditions requiring broader tissue coverage, such as ischemic areas with compromised vascular access. Conversely, in applications requiring localized angiogenic effects, the more matrix-binding VEGF164 isoform may be preferable. In experimental models of therapeutic angiogenesis, VEGF120 demonstrates potent endothelial cell proliferation and migration effects, with an ED50 of 2-10 ng/ml in HUVEC proliferation assays . The yeast-derived protein offers advantages for therapeutic applications, including high purity through proprietary chromatographic techniques and lower immunogenicity compared to proteins produced in bacterial systems . Based on studies examining VEGF isoform expression in inflammatory and regenerative contexts, the choice between VEGF120 and other isoforms should consider the temporal dynamics of the target condition. For acute therapeutic needs, the rapid diffusion and potent signaling activation of VEGF120 may be advantageous, while for sustained angiogenic support, combination with longer-acting isoforms might provide better outcomes . When designing therapeutic protocols, researchers should consider that different VEGF isoforms show distinct binding patterns with co-receptors such as neuropilins, potentially influencing vessel morphology and function in the regenerating tissue.

What considerations are important when using rat VEGF (120a.a.) in studies of pathological versus physiological angiogenesis?

When using rat VEGF (120a.a.) to study pathological versus physiological angiogenesis, several critical considerations must guide experimental design and interpretation. First, dosage calibration is essential—physiological angiogenesis typically involves coordinated, moderate VEGF expression (2-10 ng/ml in in vitro systems) , while pathological angiogenesis often features higher, sustained VEGF concentrations. Researchers should establish dose-response curves specific to their model systems to identify the threshold between physiological and pathological responses. Temporal dynamics also differ significantly—physiological angiogenesis follows precise, self-limiting kinetics, while pathological angiogenesis shows persistent activation. Implementation of inducible or pulsatile VEGF delivery systems can help model these distinct temporal patterns. The cellular context substantially influences VEGF responses; co-culture systems incorporating appropriate supporting cells (pericytes, fibroblasts, immune cells) provide more representative models than endothelial monocultures. For in vivo studies, the method of VEGF administration critically determines outcomes—controlled release formulations typically produce more organized vasculature resembling physiological angiogenesis, while bolus injections often lead to leaky, disorganized vessels characteristic of pathological conditions . Assessment metrics must extend beyond simple vessel density to include functional measures such as perfusion efficiency, vascular permeability, pericyte coverage, and vessel maturation markers. The inflammatory context also plays a decisive role—studies have shown that VEGF signaling patterns differ significantly between acute and chronic inflammatory states, with implications for vessel structure and function . Finally, researchers should consider sex-specific responses, as male and female animals show differential sensitivity to VEGF/VEGFR2 modulation, particularly in inflammatory contexts .

What are common issues encountered when using rat VEGF (120a.a.) in cell-based assays, and how can they be resolved?

Researchers frequently encounter several challenges when using rat VEGF (120a.a.) in cell-based assays, each requiring specific troubleshooting approaches. Low biological activity is a common issue that may result from protein degradation during storage or handling. To resolve this, store lyophilized VEGF at -20°C and reconstituted protein at 4°C for short-term use or -80°C in single-use aliquots for long-term storage . Verify activity using positive control cells known to respond robustly to VEGF, such as HUVECs, with an expected ED50 range of 2-10 ng/ml . Variable cellular responsiveness across experiments often stems from differences in cell passage number or culture conditions. Standardize by using low-passage cells (passages 3-4 for HUVECs), implement consistent serum starvation protocols (0.1% FBS for 24 hours) , and establish internal standards for normalization between experiments. Cross-species reactivity concerns may arise when using rat VEGF with human or other species' cells. While rat VEGF generally activates human VEGFR2, efficacy may vary; if diminished responses occur, consider species-matched VEGF or validate cross-reactivity through receptor phosphorylation assays. Background activation of VEGF pathways by serum components can mask experimental effects; minimize by reducing serum concentration during experiments and include appropriate vehicle controls. Adsorption of VEGF to plasticware reduces effective concentration; prevent by adding carrier proteins (0.1-0.5% BSA) to working solutions and using low-protein binding plasticware. For detection problems in signaling studies, optimize antibody selection—ensure antibodies recognize phosphorylated VEGFR2 and downstream targets like ERK and PLCγ with appropriate specificity . If cellular responses plateau or appear non-linear, implement broader dose ranges (0.1-100 ng/ml) to capture the full response spectrum.

How can I optimize Western blot detection of VEGF-induced signaling pathways when using rat VEGF (120a.a.)?

Optimizing Western blot detection of VEGF-induced signaling requires attention to several critical parameters throughout the experimental workflow. Begin with careful timing of VEGF stimulation—phosphorylation of VEGFR2 peaks rapidly (within 5-15 minutes) while downstream effectors like ERK1/2 and PLCγ show more variable kinetics . Implement a time-course experiment (5, 15, 30, 60 minutes) to capture the optimal activation window for each target. Cell preparation significantly impacts baseline phosphorylation; standardize by serum-starving cells (0.1% FBS in EBM) for 24 hours prior to VEGF treatment . For lysate preparation, use phosphatase inhibitor-rich lysis buffers to preserve phosphorylation status, and maintain samples at 4°C throughout processing. Protein separation quality affects resolution of phosphorylated species; employ 4-20% gradient SDS-PAGE gels for optimal separation of both high molecular weight receptors (VEGFR2 ~230 kDa) and smaller downstream effectors . During transfer, use PVDF membranes for phosphorylated proteins with extended transfer times (3 hours at 300 mA) for complete transfer of large proteins like VEGFR2 . Blocking should balance background reduction without epitope masking; 5% non-fat milk in TBST works well for many antibodies, but switch to BSA-based blocking for phospho-specific antibodies . For primary antibodies, select those validated specifically for rat VEGF-induced signaling, and optimize concentrations through titration experiments. Detection sensitivity can be enhanced using signal amplification systems appropriate for the phosphorylation status being examined. When analyzing results, always normalize phospho-protein signals to total protein levels of the same target to account for loading variations and expression differences. Finally, validate key findings with supporting approaches like phospho-ELISAs or functional assays to confirm the biological relevance of observed signaling changes.

What are the key considerations when transitioning from in vitro to in vivo studies with rat VEGF (120a.a.)?

Transitioning from in vitro to in vivo studies with rat VEGF (120a.a.) requires careful attention to multiple factors that influence experimental success and data interpretation. First, dosage scaling is critical—effective in vitro concentrations (2-10 ng/ml) cannot be directly translated to in vivo dosing due to differences in distribution volume, protein half-life, and tissue-specific uptake. Conduct pilot dose-response studies to determine effective in vivo concentrations that produce measurable biological effects without toxicity. Delivery method significantly impacts VEGF bioavailability and activity; options include bolus injection (suitable for acute effects), osmotic pumps (for sustained delivery), or matrix-bound formulations (for localized effects). Each approach yields different pharmacokinetic profiles and angiogenic outcomes. Consider strain-specific responses, as different rat strains may exhibit variable sensitivity to VEGF—reports demonstrate distinct responses in MWF rats compared to other strains . Sex-specific effects are particularly important; studies have documented differential responses to VEGF/VEGFR2 modulation between male and female rats, with greater effects observed in males during chronic inflammatory conditions . For quantitative assessment of angiogenic responses, implement complementary methodologies: confocal microscopy with fluorescent vascular labeling (using Alexa Fluor® 647 WGA) enables three-dimensional visualization and quantification of vessel networks , while immunohistochemistry for VEGF and related factors provides molecular context . Control for confounding variables including age (which affects vascular responsiveness), concurrent medications, and comorbid conditions that may alter VEGF signaling. Finally, experimental duration must align with the vascular processes being studied—sprouting angiogenesis begins within days, while vessel maturation and remodeling require weeks to assess properly. To establish causality, incorporate VEGFR2 inhibitors (such as Ki-8751) as experimental controls to confirm VEGF-specific effects .

How might the study of rat VEGF (120a.a.) contribute to understanding the role of different VEGF isoforms in tissue-specific angiogenesis?

The study of rat VEGF (120a.a.) presents valuable opportunities for elucidating isoform-specific functions in tissue-specific angiogenesis. Unlike larger isoforms that contain heparin-binding domains, VEGF120 exhibits greater diffusibility, making it particularly informative for understanding how soluble versus matrix-bound VEGF gradients differentially regulate vascular patterning . Future research should employ parallel experimental platforms comparing VEGF120 with other isoforms (VEGF164, VEGF188) across multiple tissue environments to develop comprehensive models of isoform-specific angiogenic regulation. Comparative studies examining the molecular interactome of VEGF120 versus other isoforms could identify unique binding partners that mediate tissue-specific effects. The development of isoform-specific neutralizing antibodies or inhibitors would enable precise manipulation of individual VEGF variants without affecting others, allowing for targeted intervention approaches . Studies examining differential expression of VEGF120 versus other isoforms across tissues and during various physiological and pathological conditions would provide insight into their specialized roles—preliminary evidence suggests tissue-specific distribution patterns of VEGF-A isoforms (VEGF-Axxxa/VEGF-Axxxb) that change during inflammation . Advanced imaging techniques combining isoform-specific labeling with functional vascular assessment could map the spatiotemporal relationship between specific VEGF variants and resulting vascular structures. Additionally, genetic approaches using conditional, isoform-specific knockouts would reveal the necessity of VEGF120 for particular aspects of physiological or pathological angiogenesis in specific tissues. This multifaceted approach would significantly advance our understanding of how the complex VEGF isoform system achieves precise tissue-specific vascular regulation.

What emerging technologies might enhance the therapeutic applications of rat VEGF (120a.a.) in regenerative medicine?

Emerging technologies are poised to revolutionize the therapeutic applications of rat VEGF (120a.a.) in regenerative medicine through several innovative approaches. Advanced delivery systems using biodegradable polymers or hydrogels could provide spatiotemporally controlled release of VEGF120, mimicking the precise regulation observed in physiological angiogenesis . These smart materials could respond to tissue microenvironment cues such as pH, matrix metalloproteinases, or hypoxia to adjust VEGF release rates based on regenerative needs. Combination delivery platforms incorporating VEGF120 with complementary factors like FGF2, HGF, and IGF-1 may enhance therapeutic outcomes by recapitulating the growth factor milieu observed in successful regenerative processes . Gene editing technologies could enable targeted modification of endogenous VEGF expression patterns to favor specific isoforms in particular contexts, potentially shifting the VEGF-Axxxa/VEGF-Axxxb balance toward regenerative phenotypes . Cell-based approaches using stem cells or progenitors genetically modified to express optimal levels of VEGF120 under regulated conditions may provide more physiological delivery than bolus protein administration. Organ-on-chip technologies incorporating VEGF120 in microfluidic systems could serve as personalized testing platforms to optimize dosing and combination strategies before in vivo application. Nanobody or aptamer-based technologies targeting VEGF receptors could provide more selective activation of beneficial VEGF signaling pathways while minimizing unwanted effects. Injectable, in situ polymerizing hydrogels containing VEGF120 and adhesion motifs could create artificial niches supporting tissue-specific regeneration. Additionally, the application of machine learning algorithms to predict optimal VEGF120 dosing regimens based on patient-specific factors, including sex differences in VEGF responsiveness , could lead to more personalized and effective regenerative approaches.

How can computational modeling informed by experimental data with rat VEGF (120a.a.) improve our understanding of angiogenic network regulation?

Computational modeling informed by experimental data with rat VEGF (120a.a.) offers tremendous potential for advancing our understanding of angiogenic network regulation. Agent-based models simulating endothelial cell behaviors (proliferation, migration, and tube formation) in response to VEGF120 gradients could predict emergent vascular patterns under various conditions. These models can incorporate experimentally derived parameters such as the ED50 values (2-10 ng/ml) and temporal dynamics of VEGF-induced signaling to ensure biological relevance. Multi-scale models integrating molecular signaling networks (VEGFR2, ERK1/2, PLCγ phosphorylation cascades) with cellular behaviors and tissue-level outcomes would bridge the gap between molecular mechanisms and physiological effects. Pharmacokinetic/pharmacodynamic (PK/PD) modeling could optimize dosing regimens by predicting VEGF120 distribution, receptor occupancy, and resulting angiogenic responses across different tissues and time scales. Network analysis approaches connecting VEGF signaling with other growth factor pathways (FGF2, HGF, IGF-1) would map the complex interactome regulating angiogenesis in different physiological and pathological contexts. Sex-specific modeling frameworks incorporating the differential responses observed between male and female subjects could elucidate the mechanisms underlying these differences and inform sex-specific therapeutic approaches. Machine learning algorithms trained on experimental datasets could identify previously unrecognized patterns in VEGF-responsive gene expression or cellular behaviors, generating novel hypotheses for experimental validation. In silico screening of VEGFR2 modulators based on structural data could accelerate the discovery of compounds with optimal efficacy and specificity profiles. Systems biology approaches integrating transcriptomic, proteomic, and metabolomic data from VEGF120-treated systems would provide comprehensive maps of the angiogenic response network. Importantly, these computational approaches should maintain tight integration with experimental validation studies to ensure biological relevance and continually refine model parameters based on new experimental findings.

Product Science Overview

Introduction

Vascular Endothelial Growth Factor (VEGF) is a critical signaling protein involved in both angiogenesis (the formation of new blood vessels from pre-existing vessels) and vasculogenesis (the formation of entirely new blood vessels during embryonic development). VEGF plays a pivotal role in the body’s ability to supply oxygen and nutrients to tissues by promoting the growth of blood vessels. This protein is particularly important in conditions where tissues are deprived of oxygen, such as in wound healing and the growth of tumors.

Structure and Composition

The specific form of VEGF in question, Vascular Endothelial Growth Factor (120a.a.) (Rat Recombinant, Yeast), is a recombinant protein produced in yeast. It consists of 120 amino acids and is a disulfide-linked homodimer, meaning it is composed of two identical polypeptide chains linked by disulfide bonds. Each polypeptide chain has a molecular mass of approximately 18.5 kDa .

Production and Purification

This recombinant form of VEGF is produced using the yeast expression system, specifically Saccharomyces cerevisiae. The yeast system is often used for the production of recombinant proteins due to its ability to perform post-translational modifications and its ease of scaling up for industrial production. The VEGF protein is purified using proprietary chromatographic techniques to ensure high purity and activity .

Biological Activity

VEGF is known for its ability to stimulate endothelial cell mitogenesis (cell division) and cell migration. It also increases vascular permeability and acts as a vasodilator. These properties make VEGF a key player in the formation of new blood vessels. In vitro studies have shown that VEGF can stimulate the proliferation of human umbilical vein endothelial cells (HUVECs) at concentrations as low as 2-10 ng/ml .

Clinical Significance

Elevated levels of VEGF have been associated with various pathological conditions, including cancer, diabetic retinopathy, and POEMS syndrome (a rare blood disorder). In cancer, VEGF promotes tumor growth by ensuring an adequate blood supply to the rapidly dividing cancer cells. As a result, VEGF has become a target for anti-cancer therapies aimed at inhibiting its activity and thereby restricting the blood supply to tumors .

Applications

Recombinant VEGF proteins, such as the 120 amino acid form from rats, are used extensively in research to study angiogenesis and related processes. They are also used in the development of therapeutic agents aimed at modulating angiogenesis in various diseases. For example, VEGF inhibitors are used in the treatment of certain cancers and eye diseases like age-related macular degeneration.

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.