Feature | Human VEGF-D | Mouse Homolog |
---|---|---|
Alternative name | c-Fos-induced growth factor | FIGF |
Receptor binding | VEGFR-2 & VEGFR-3 | VEGFR-3 dominant |
Amino acid identity | 100% | 94% |
Receptor | Binding Affinity | Primary Signaling Outcome |
---|---|---|
VEGFR-2 | High (Kd 135 pM) | Angiogenesis, vascular permeability |
VEGFR-3 | Moderate | Lymphangiogenesis, vessel maturation |
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:
Gene therapy applications:
Lymphatic disorders: Serum levels >800 pg/mL diagnostic for lymphangioleiomyomatosis
Cancer progression:
Parameter | VEGF-D | VEGF-C | VEGF-A |
---|---|---|---|
Receptor specificity | VEGFR-2/3 | VEGFR-2/3 | VEGFR-1/2 |
Lymphangiogenic potency | ++++ | +++ | + |
Angiogenic index (vs control) | 4.2x | 1.8x | 3.9x |
Plasma half-life | 15-20 min | 10-15 min | 3-6 min |
Neutralizing antibodies (e.g., V0885):
mTOR inhibitors:
Gene therapy vectors:
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 .
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 .
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 .
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
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 .
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 .
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 .
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 .
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:
Pharmacodynamic biomarkers:
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 Type | Markers | Clinical Application | Sample Type |
---|---|---|---|
Protein Level | Circulating VEGF-D | LAM diagnosis, tumor burden | Serum/plasma |
Tissue Expression | VEGF-D immunohistochemistry | Vascular phenotyping | Tumor biopsies |
Genetic Signature | VDV gene expression | Anti-VEGF therapy response prediction | Pretreatment tumor tissue |
Receptor Activation | VEGFR-2/3 phosphorylation | Pathway activity assessment | Fresh tissue samples |
Downstream Targets | Endothelial response genes | Pharmacodynamic monitoring | Tumor biopsies |
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:
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:
Therapeutic delivery:
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 .
Several promising research directions for VEGF-D are emerging:
Personalized medicine approaches:
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:
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" .
Looking forward, several developments may transform how VEGF-D-related research impacts clinical care:
Biomarker-guided therapy selection:
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
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.
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 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.
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 .
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 .