VEGF D Human

Vascular Endothelial Growth Factor D Human Recombinant
Shipped with Ice Packs
In Stock

Description

Genetic and Species Variation

FeatureHuman VEGF-DMouse Homolog
Alternative namec-Fos-induced growth factorFIGF
Receptor bindingVEGFR-2 & VEGFR-3VEGFR-3 dominant
Amino acid identity100%94%

Receptor Activation Profile

ReceptorBinding AffinityPrimary Signaling Outcome
VEGFR-2High (Kd 135 pM)Angiogenesis, vascular permeability
VEGFR-3ModerateLymphangiogenesis, vessel maturation

Key Physiological Roles:

  • Developmental: Critical for neonatal lymphatic maturation and bone growth

  • Adult tissue maintenance: Expressed in heart (25% of total), lung (18%), skeletal muscle (15%), and intestinal tissues

  • Pathological involvement:

    • Tumor lymphangiogenesis and metastasis

    • Cardiac remodeling post-myocardial infarction

    • Lymphangioleiomyomatosis progression

Therapeutic Potential

  • Gene therapy applications:

    • Adenoviral VEGF-DΔNΔC induces 3.8-fold greater capillary enlargement vs VEGF-A

    • Full-length VEGF-D shows 2.1-fold higher lymphangiogenic activity than VEGF-C

Diagnostic Biomarker Performance

Clinical ContextVEGF-D AssociationHazard Ratio (95% CI)
All-cause mortalityPositive correlation1.32 (1.12-1.56) per log SD
Cardiovascular deathStrong association1.41 (1.14-1.74)
Major adverse eventsNo significant link1.07 (0.90-1.27)

Disease Correlations

  • Lymphatic disorders: Serum levels >800 pg/mL diagnostic for lymphangioleiomyomatosis

  • Cancer progression:

    • Ovarian cancer: SPARC-mediated VEGF-D inhibition reduces metastasis

    • Melanoma: VEGF-D promotes sentinel lymph node invasion

Comparative Analysis with VEGF Family Members

ParameterVEGF-DVEGF-CVEGF-A
Receptor specificityVEGFR-2/3VEGFR-2/3VEGFR-1/2
Lymphangiogenic potency++++++++
Angiogenic index (vs control)4.2x1.8x3.9x
Plasma half-life15-20 min10-15 min3-6 min

Current Approaches:

  1. Neutralizing antibodies (e.g., V0885):

    • Cross-reactivity: 25% with mouse VEGF-D, none with human VEGF/VEGF-B

    • IC₅₀: 0.5-2.0 μg/mL in endothelial proliferation assays

  2. mTOR inhibitors:

    • Rapamycin reduces serum VEGF-D by 43% in LAM patients (P<0.001)

  3. Gene therapy vectors:

    • Adenoviral delivery achieves 14-day sustained expression in muscle tissue

Product Specs

Introduction
VEGF-D, a member of the VEGF/PDGF protein family, plays a crucial role in promoting lymphangiogenesis and endothelial cell growth. It also contributes to regulating vascular permeability. Moreover, VEGF-D is essential for developing the venous and lymphatic vascular systems, along with the growth and maintenance of differentiated lymphatic endothelium. In its mature form, VEGF-D exists as a noncovalently linked homodimer, binding to and activating both VEGFR-2 (flk1) and VEGFR-3 (flt4).
Description
Recombinant human VEGFD, produced in HEK-293 cells, is a secreted protein encompassing amino acids Phe93 to Ser201. It is fused to a polyhistidine tag at the C-terminus.
Physical Appearance
Sterile Filtered White lyophilized powder.
Formulation
Following extensive dialysis against PBS, the recombinant VEGF-D was lyophilized.
Solubility
For reconstitution, it is advised to dissolve the Vascular Endothelial Growth Factor D in sterile 18M-cm H₂O at a concentration of at least 100 µg/ml. This solution can then be further diluted with other aqueous solutions as required.
Stability
While lyophilized VEGF-D remains stable at room temperature for up to 3 weeks, it is recommended to store it desiccated below -18°C. After reconstitution, VEGF-D should be kept at 4°C for 2-7 days. For extended storage, freezing below -18°C is recommended. To enhance long-term stability during storage, adding a carrier protein (0.1% HSA or BSA) is advisable. Avoid repeated freeze-thaw cycles.
Purity
Purity exceeding 95.0% as determined by SDS-PAGE analysis.
Biological Activity
The biological activity, represented by ED50, is determined to be 3-4 ng/ml. This value is measured based on the protein's ability to stimulate the proliferation of human microvascular endothelial cells (HMVECs).
Synonyms
c-fos induced growth factor (vascular endothelial growth factor D), FIGF, VEGFD.
Source
HEK293.

Q&A

What is VEGF-D and how does it differ from other VEGF family members?

VEGF-D, also known as c-Fos-induced growth factor (FIGF), is a member of the PDGF/VEGF growth factor family with distinct expression patterns and receptor binding properties. Unlike other VEGF family members, VEGF-D shows particularly high expression in lung, heart, and small intestine tissues, with lower levels in skeletal muscle, colon, and pancreas. It has the distinctive ability to bind both VEGFR-2 and VEGFR-3 receptors, making it uniquely positioned to influence both blood vessel angiogenesis and lymphatic vessel development simultaneously .

The protein structure of VEGF-D includes N- and C-terminal propeptides that undergo proteolytic processing to generate mature forms with enhanced receptor binding affinity. This post-translational processing represents a key regulatory mechanism that distinguishes VEGF-D from some other family members and influences its biological activities in different tissue contexts .

What are the primary signaling pathways activated by VEGF-D?

VEGF-D activates multiple downstream signaling cascades through binding to VEGFR-2 and VEGFR-3 receptors. Upon receptor binding, VEGF-D triggers receptor dimerization and autophosphorylation, leading to activation of several pathways including:

  • PI3K/Akt pathway - promoting endothelial cell survival

  • MAPK/ERK pathway - stimulating proliferation

  • p38 MAPK pathway - influencing cell migration

  • PLCγ/PKC pathway - affecting vascular permeability

The specific biological outcomes depend on receptor expression patterns, co-receptor availability, and the cellular context. Research indicates that VEGF-D dimers demonstrate enhanced bioactivity compared to monomeric forms, with implications for physiological functions and pathological processes .

What are the optimal approaches for studying VEGF-D expression and activity in experimental models?

Multiple complementary approaches are recommended for comprehensive VEGF-D analysis:

  • Gene expression analysis:

    • qRT-PCR for mRNA quantification

    • RNA-seq for transcriptome-wide context

    • In situ hybridization for spatial localization

  • Protein detection methods:

    • ELISA for quantitative measurement in tissue lysates or blood

    • Western blotting for processing forms analysis

    • Immunohistochemistry for tissue localization

  • Functional assays:

    • Endothelial tube formation assays

    • Migration/chemotaxis assays

    • Receptor phosphorylation assessment

For in vivo studies, compartment models have proven valuable, particularly for studying VEGF-D dynamics in tumor microenvironments. These models can simulate complex interactions between normal tissue, blood, and tumor compartments, providing insights difficult to obtain through direct measurement .

How can researchers accurately model VEGF-D dynamics in tumor systems?

Computational modeling approaches have emerged as powerful tools for understanding VEGF-D behavior in complex biological systems. Three-compartment models that simulate VEGF transport and kinetics between normal tissue, blood, and tumor compartments have been successfully developed.

These models can predict parameters that are challenging to measure experimentally, such as:

  • VEGF secretion rates from different cell types

  • Receptor occupancy in different tissue compartments

  • Effects of VEGF-targeting agents

For example, researchers have used such models to predict that human tumors secrete VEGF at rates ranging from 0.007 to 0.023 molecules/cell/second, which is lower than many in vitro measurements (0.03-2.65 molecules/cell/second). This highlights the importance of validating in vitro findings with in vivo models .

When designing VEGF-D studies, researchers should consider:

  • Species-specific differences in VEGF isoforms (human VEGF-D vs. mouse VEGF-D)

  • Differences between xenograft and syngeneic tumor models

  • The impact of tumor microenvironment on VEGF dynamics

How does VEGF-D contribute to cancer progression and metastasis?

VEGF-D plays multifaceted roles in cancer progression through several mechanisms:

  • Lymphangiogenesis promotion: VEGF-D stimulates the formation of new lymphatic vessels, providing routes for tumor cell dissemination. This occurs through binding to VEGFR-3 receptors on lymphatic endothelial cells, promoting their proliferation, migration, and tube formation .

  • Lymph node metastasis: Increased VEGF-D expression correlates with lymph node metastasis in multiple cancer types. VEGF-D facilitates the restructuring of lymphatic channels, enhancing the transport of tumor cells to regional lymph nodes .

  • Tumor angiogenesis: Through its interaction with VEGFR-2, VEGF-D can promote tumor blood vessel formation, supporting tumor growth and potentially hematogenous metastasis .

  • Immunomodulatory effects: Emerging evidence suggests VEGF-D may influence the tumor immune microenvironment, potentially affecting immunotherapy responses.

Research investigating VEGF-D's prognostic significance has indicated that tumors with higher VEGF-dependent vasculature (VDV) gene expression may show improved responses to anti-VEGF therapies, suggesting potential predictive biomarker applications .

What role does VEGF-D play in non-oncological diseases?

Beyond cancer, VEGF-D has been implicated in several other pathological conditions:

  • Cardiovascular diseases: VEGF-D has shown promise in therapeutic angiogenesis for conditions like refractory angina. Clinical trials using adenoviral VEGF-D gene delivery have demonstrated safety and tolerability in these patients .

  • Lymphangioleiomyomatosis (LAM): VEGF-D plays a significant role in the pathogenesis of this devastating pulmonary condition. Elevated VEGF-D levels serve as a diagnostic biomarker for LAM and may contribute to its pathological vascular remodeling .

  • Pulmonary vasculopathy: Mutations in the VEGF-D gene have been associated with distended pulmonary arteries and veins, leading to aberrant angiogenesis through increased VEGFR-2 signaling .

  • Hyperoxic acute lung injury (HALI): VEGF-D has been shown to promote pulmonary edema in mouse models of this condition, which can occur when patients receive high oxygen concentrations during ventilation .

  • Peripheral vascular disease: Preclinical studies suggest VEGF-D has therapeutic potential for improving perfusion in ischemic tissues .

What approaches have been developed for therapeutic targeting or delivery of VEGF-D?

Multiple strategies have been explored for modulating VEGF-D activity in various disease contexts:

  • VEGF-D inhibition approaches:

    • Monoclonal antibodies against VEGF-D

    • Receptor tyrosine kinase inhibitors targeting VEGFR-2/3

    • Soluble receptor traps (e.g., VEGF Trap/aflibercept)

    • RNA interference technologies

  • VEGF-D delivery for therapeutic angiogenesis:

    • Adenoviral gene transfer (most clinically advanced)

    • Plasmid-based gene therapy

    • Recombinant protein delivery

    • Biomaterial-based controlled release systems

Clinical trials have assessed adenoviral delivery of mature VEGF-D for treating refractory angina, with results indicating safety, feasibility and good tolerability. This approach has shown promise in promoting transmural angiogenesis, particularly pronounced in the epicardium .

Compartment modeling studies have provided insights into optimizing dosing of VEGF-targeted therapies. For instance, mathematical models predict that at higher doses of VEGF Trap (aflibercept), all human VEGF originating from tumors can be neutralized, providing a rational basis for dose selection in clinical studies .

How effective are VEGF-D-targeted therapies in different disease contexts?

The efficacy of VEGF-D-targeted approaches varies by disease context:

  • Cancer: Anti-VEGF therapies that affect VEGF-D signaling, such as receptor tyrosine kinase inhibitors or VEGF Trap, have shown variable efficacy. Recent studies indicate that pretreatment tumor VEGF-dependent vasculature (VDV) gene expression may predict response to bevacizumab-containing therapies .

  • Cardiovascular disease: Gene therapy delivering mature VEGF-D has shown promise in preclinical models and early-phase clinical trials for refractory angina, peripheral vascular disease, and vascular restenosis .

  • Pulmonary conditions: While therapeutic targeting of VEGF-D pathways for LAM remains experimental, VEGF-D levels have become important biomarkers for diagnosis .

A retrospective analysis of clinical trial data revealed that patients with higher expression of VEGF-dependent vasculature (VDV) genes in pretreatment tumor samples exhibited improved clinical outcomes when treated with bevacizumab-containing therapies. This suggests that VEGF bioactivity levels before treatment may predict therapeutic efficacy .

How can VEGF-D and its downstream markers be used as biomarkers in disease diagnosis or treatment response?

VEGF-D and its associated downstream markers have emerging utility as biomarkers in several contexts:

  • Diagnostic biomarkers:

    • Serum VEGF-D levels are used clinically for LAM diagnosis

    • Tissue VEGF-D expression patterns may help distinguish vascular abnormalities

  • Predictive biomarkers for therapy response:

    • VEGF-dependent vasculature (VDV) gene signatures have shown potential in predicting response to anti-VEGF therapies

    • Higher pretreatment VDV expression correlates with improved clinical outcomes in patients receiving bevacizumab-containing treatments

  • Pharmacodynamic biomarkers:

    • VEGF-dependent genes enable detection of VEGF signaling inhibition in both preclinical models and human tumor biopsies

    • These markers include direct and distal VEGF downstream endothelial targets

Research has identified surrogate markers of VEGF inhibition by profiling gene expression changes in response to VEGF blockade. These VEGF-dependent vasculature (VDV) genes can detect VEGF signaling inhibition and potentially predict therapeutic response .

The following table summarizes key VEGF-D-related biomarker approaches:

Biomarker TypeMarkersClinical ApplicationSample Type
Protein LevelCirculating VEGF-DLAM diagnosis, tumor burdenSerum/plasma
Tissue ExpressionVEGF-D immunohistochemistryVascular phenotypingTumor biopsies
Genetic SignatureVDV gene expressionAnti-VEGF therapy response predictionPretreatment tumor tissue
Receptor ActivationVEGFR-2/3 phosphorylationPathway activity assessmentFresh tissue samples
Downstream TargetsEndothelial response genesPharmacodynamic monitoringTumor biopsies

What are the major technical challenges in VEGF-D research and how can they be addressed?

Researchers face several technical challenges when studying VEGF-D:

  • Protein detection specificity:

    • Challenge: Distinguishing between different processed forms of VEGF-D

    • Solution: Use antibodies specific to particular domains or processed forms; employ mass spectrometry for detailed characterization

  • Model system limitations:

    • Challenge: Species differences in VEGF biology between humans and rodent models

    • Solution: Use humanized mouse models; employ computational approaches like compartment modeling to integrate across species

  • Functional assessment:

    • Challenge: Isolating VEGF-D-specific effects from redundant VEGF family activities

    • Solution: Use receptor-specific blocking antibodies; develop VEGF-D knockout and knockin models with specific mutations

  • Downstream signaling complexity:

    • Challenge: Identifying VEGF-D-specific gene signatures amid complex angiogenic pathways

    • Solution: Apply systems biology approaches and carefully designed VEGF-D perturbation studies to identify direct targets

  • Therapeutic delivery:

    • Challenge: Achieving sustained local delivery of VEGF-D for therapeutic angiogenesis

    • Solution: Develop controlled release systems; optimize viral vector design for gene therapy approaches

Compartment modeling approaches have proven valuable in addressing some of these challenges by enabling researchers to predict parameters that are difficult to measure experimentally, such as VEGF secretion rates and distribution across different tissue compartments .

What are the emerging areas of VEGF-D research with high potential impact?

Several promising research directions for VEGF-D are emerging:

  • Personalized medicine approaches:

    • Validating VDV gene signatures as predictive biomarkers in prospective clinical trials

    • Developing patient selection strategies based on tumor VEGF-D pathway activity

  • Novel delivery technologies:

    • Nanoparticle-based delivery of VEGF-D or inhibitors

    • Tissue-specific gene therapy approaches with enhanced safety profiles

    • Smart delivery systems responsive to hypoxia or other microenvironmental cues

  • Combination therapy strategies:

    • Combining VEGF-D-targeted approaches with immune checkpoint inhibitors

    • Dual targeting of lymphangiogenic and angiogenic pathways

    • Sequential therapy approaches to overcome resistance mechanisms

  • Expanded disease applications:

    • Exploring VEGF-D roles in neurological disorders

    • Investigating lymphatic contributions to inflammatory diseases

    • Developing VEGF-D-based approaches for lymphedema and other lymphatic disorders

  • Advanced computational modeling:

    • Integrating multi-scale models from molecular interactions to tissue-level effects

    • Developing patient-specific models for predicting therapy response

    • Combining imaging data with computational approaches for real-time therapy optimization

The integration of computational modeling, systems biology approaches, and biomarker-guided therapy represents a particularly promising direction. As noted in the research literature, "The compartment model presented here provides a framework to investigate the action of VEGF-targeting agents for particular types of tumors... and may be useful in evaluating biomarkers of anti-angiogenic therapies" .

How might advances in targeting VEGF-D impact clinical practice in the next decade?

Looking forward, several developments may transform how VEGF-D-related research impacts clinical care:

  • Biomarker-guided therapy selection:

    • Implementation of VDV gene expression analysis in routine clinical testing

    • Development of companion diagnostics for anti-angiogenic therapies

    • Real-time monitoring of VEGF pathway activity during treatment

  • Expanded therapeutic applications:

    • Approval of VEGF-D gene therapy for cardiovascular indications

    • Novel applications in lymphatic disorders and inflammatory diseases

    • Combination regimens incorporating VEGF-D targeting with other modalities

  • Precision medicine approaches:

    • Patient-specific dosing based on computational modeling

    • Genomic and proteomic profiling to predict VEGF-D pathway dependencies

    • Adaptive therapy approaches based on dynamic biomarkers

  • Technological innovations:

    • Implantable devices for controlled VEGF-D delivery

    • Minimally invasive imaging of VEGF-D activity

    • AI-assisted interpretation of complex VEGF pathway biomarkers

Product Science Overview

Introduction

Vascular Endothelial Growth Factor D (VEGF-D) is a member of the VEGF family, which plays a crucial role in angiogenesis and lymphangiogenesis. VEGF-D is a protein encoded by the VEGFD gene and is involved in the formation of blood and lymphatic vessels. This article delves into the structure, function, and significance of VEGF-D, particularly focusing on its recombinant form used in research and therapeutic applications.

Structure and Function

VEGF-D binds to and induces dimerization and tyrosine autophosphorylation of its endothelial cell-specific receptors, VEGFR-2 and VEGFR-3 . VEGFR-2 signals stimulate endothelial sprouting, proliferation, and survival, as well as vascular permeability. VEGFR-3 signals stimulate similar processes in lymphatic endothelial cells . Unlike VEGF-A and VEGF-C, which are indispensable for embryonic vascular development, VEGF-D can be deleted without any obvious phenotype . However, recombinant VEGF-D is capable of inducing angiogenesis and lymphangiogenesis in several experimental conditions, suggesting its potential therapeutic utility in regenerative medicine .

Recombinant VEGF-D

Recombinant human VEGF-D is produced using various expression systems, including bacterial, yeast, and mammalian cells. The recombinant form is used extensively in research to study its biological activities and potential therapeutic applications. The production process typically involves cloning the VEGFD gene into an expression vector, transforming the host cells, and purifying the expressed protein.

Biological Activities

VEGF-D has been shown to promote the growth of blood and lymphatic vessels, making it a key player in both physiological and pathological angiogenesis. It is involved in processes such as wound healing, tissue repair, and tumor growth. VEGF-D’s ability to induce angiogenesis and lymphangiogenesis makes it a potential target for therapeutic interventions in diseases characterized by abnormal vessel growth, such as cancer and lymphedema .

Therapeutic Applications

The therapeutic potential of VEGF-D lies in its ability to modulate angiogenesis and lymphangiogenesis. In regenerative medicine, recombinant VEGF-D could be used to promote tissue repair and regeneration. Additionally, VEGF-D-targeted therapies are being explored for their potential to inhibit tumor growth by blocking the formation of new blood vessels that supply nutrients to tumors .

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.