Platelet-derived growth factor D (PDGFD) is a homodimeric protein encoded by the PDGFD gene (chromosome 11q22.3) and belongs to the PDGF family, which includes PDGF-A, PDGF-B, PDGF-C, and PDGF-D . It plays critical roles in embryonic development, tissue repair, and pathological processes such as cancer progression and fibrosis. Below is a detailed analysis of its molecular characteristics, biological functions, and clinical implications.
Protein:
Parameter | Value | Source |
---|---|---|
Molecular weight | 42.8 kDa (mature) | |
Theoretical pI | 8.04 | |
Amino acid sequence | 370 residues (precursor) | |
Chromosomal location | 11q22.3 |
PDGFD binds to PDGFR-β, inducing receptor homodimerization and activating downstream signaling (e.g., PI3K/Akt, MAPK/ERK pathways) . It may also form heterodimers with PDGFR-α in certain contexts .
Receptor | Interaction | Outcome |
---|---|---|
PDGFR-β | Homodimerization | Cell proliferation, migration |
PDGFR-α | Heterodimerization (rare) | Complementary signaling |
Embryonic development: Critical for mesenchymal cell migration, neural crest patterning, and organogenesis .
Tissue repair: Promotes wound healing by recruiting macrophages and inducing angiogenesis .
Cellular maintenance: Supports survival and chemotaxis of mesenchymal cells (e.g., smooth muscle, fibroblasts) .
PDGFD overexpression is implicated in:
Disease | Mechanism | Evidence |
---|---|---|
Cancers | EMT, angiogenesis, metastasis | Colorectal, breast, renal |
Fibrosis | Extracellular matrix deposition | Renal, hepatic, cardiac |
Vascular diseases | Atherosclerosis, pulmonary hypertension | Smooth muscle cell proliferation |
Glomerulonephritis | Macrophage infiltration | Mesangial cell activation |
Colorectal cancer (CRC): PDGFD upregulates Twist1 and MMP9, driving epithelial-mesenchymal transition (EMT) .
Breast cancer: Enhances CXCR4 expression, promoting lymph node metastasis .
Angiogenesis: Induces VEGF and MMP9 expression, supporting tumor neovascularization .
Antibodies:
Tyrosine kinase inhibitors:
Agent | Target | Outcome |
---|---|---|
MAB1159 | PDGFD | Inhibits cell proliferation |
Imatinib | PDGFR-β | Reduces angiogenesis |
Dual roles: PDGFD enhances NK cell survival via PDGFR-β (beneficial for immunotherapy) but promotes tumor growth .
Resistance: Overexpression in tumors may limit therapeutic efficacy .
Human PDGF-D consists of an N-terminal CUB (complement subcomponents C1r/C1s, Uegf, and Bmp1) domain and a C-terminal PDGF/vascular endothelial growth factor domain . Unlike traditional PDGF family members (PDGF-A and PDGF-B), PDGF-D requires proteolytic removal of its CUB domain to activate PDGF receptors . The protein forms disulfide-bonded homodimers but does not heterodimerize with PDGF-A or PDGF-B chains . The recombinant form used in many laboratory applications comprises amino acids Asp253-Arg370 with accession number Q9GZP0 .
PDGF-D, also named spinal cord-derived growth factor B (SCDGF-B), differs from classical PDGF family members in several key ways . Unlike PDGF-A and PDGF-B which are secreted in active forms, PDGF-D contains an N-terminal CUB domain that requires proteolytic cleavage for receptor activation . Additionally, while PDGF-A and PDGF-B can form both homodimers and heterodimers, PDGF-D (like PDGF-C) only forms homodimers . These structural differences impact receptor specificity, with PDGF-D primarily signaling through the PDGF-β receptor rather than through multiple receptor combinations .
PDGF-D is secreted as a latent form that requires extracellular proteolytic processing to become biologically active . The processing involves removal of the N-terminal CUB domain, which exposes the PDGF/VEGF domain needed for receptor binding and activation . Research in prostate cancer cell lines has demonstrated that some cells, such as LNCaP cells, can auto-activate latent PDGF-D into its active form, which then induces phosphorylation of the β-PDGF receptor . This activation mechanism represents an important regulatory step in PDGF-D signaling that distinguishes it from other growth factors that are secreted in active forms.
PDGF-D expression is regulated by several genetic factors, including transcription factors FOXC1 and FOXC2 . The SNP rs2019090 at the 11q22.3 locus significantly influences PDGF-D expression levels . This polymorphism is multi-allelic with A as the reference allele and T, C, and G as alternatives . Position weight matrix analyses show that the T allele decreases FOXC1/C2 binding affinity to the regulatory region, resulting in reduced PDGF-D expression . CRISPR interference (CRISPRi) experiments utilizing dCas9KRAB with specific guide RNAs have confirmed the regulatory relationship between these factors and PDGF-D expression .
Genetic variations in PDGF-D have been associated with phenotypic differences in animal models, particularly in sheep tail morphology . Analysis of polymorphisms like g.4122606 C>G and g.3852134 C>T has revealed significant associations with tail measurements . For example, sheep with the GG genotype at g.4122606 C>G showed significantly smaller tail width (19.168 ± 0.204 cm) compared to those with the CC genotype (19.940 ± 0.276 cm) . Similarly, the TT genotype at g.3852134 C>T was associated with significantly larger tail length measurements (12.996 ± 1.382 cm) compared to the CC genotype (9.657 ± 0.089 cm) . These variations affect transcription factor binding in the PDGF-D promoter region, potentially altering gene expression and subsequent developmental outcomes.
Several experimental approaches have proven effective for manipulating PDGF-D expression:
Viral vector systems: Lentiviral vectors carrying PDGF-D have been successfully used for overexpression in cell culture models, such as preadipocytes . Adenoviral constructs encoding PDGF-D are effective for in vivo delivery in animal models .
Gene silencing: CRISPRi with dCas9KRAB and specific guide RNAs has effectively suppressed PDGF-D expression in human coronary artery smooth muscle cells (HCASMC) .
Tissue-specific expression: AAV vectors with tissue-specific promoters (such as RPE-specific VDM2 promoter) can drive localized PDGF-D expression in target tissues .
Transgenic models: Mice with PDGF-D expressed in basal epidermal cells have been created to study its role in skin biology and wound healing .
When implementing these approaches, researchers should consider target cell type, duration of expression required, and potential off-target effects that may influence experimental outcomes.
PDGF-D exerts multiple cellular effects through activation of PDGF receptors, primarily PDGFR-β . Key cellular responses include:
Mitogenic activity: PDGF-D is a potent stimulator of cell proliferation in mesangial cells, smooth muscle cells, and fibroblasts .
Chemotactic effects: PDGF-D induces cell migration, particularly in fibroblasts and macrophages .
Maturation of blood vessels: PDGF-D promotes pericyte/smooth muscle cell coating of blood vessels during angiogenesis .
Regulation of interstitial fluid pressure: Transgenic mice expressing PDGF-D show elevated interstitial fluid pressure in the dermis .
Adipocyte differentiation: PDGF-D appears to influence adipogenesis based on oil red O staining experiments in cell culture models .
These cellular effects are mediated through receptor-induced signaling cascades that involve receptor autophosphorylation and activation of downstream pathways.
PDGF-D works cooperatively with other growth factors during angiogenesis. When co-expressed with vascular endothelial growth factor-E (VEGF-E), PDGF-D significantly enhances pericyte/smooth muscle cell recruitment to newly formed blood vessels . This interaction leads to increased maturation of VEGF-E-induced vessels and inhibits the vascular leakiness that typically accompanies VEGF-E-induced angiogenesis . The complementary roles of these growth factors highlight the complex coordination required for proper blood vessel formation: VEGF primarily stimulates endothelial cell proliferation and initial vessel formation, while PDGF-D promotes the recruitment of supporting cells necessary for vessel stabilization and maturation.
PDGF-D plays a significant role in tissue repair and wound healing processes. In transgenic mice expressing PDGF-D in basal epidermal cells, wound healing is characterized by increased cell density and enhanced recruitment of macrophages to the healing site . The macrophage recruitment function appears to be a consistent effect of PDGF-D, as similar responses were observed when PDGF-D was expressed in skeletal muscle or ear tissues using adeno-associated virus vectors . Beyond macrophage recruitment, PDGF-D likely contributes to wound healing through its mitogenic effects on fibroblasts and its role in supporting proper vascular remodeling during the repair process.
PDGF-D has been implicated as a significant factor in coronary artery disease (CAD) risk through genome-wide association studies that identified the 11q22.3 locus containing the PDGF-D gene . Mechanistic studies have verified that the rs2019090 polymorphism at this locus affects PDGF-D expression, with the T allele associated with decreased FOXC1/C2 transcription factor binding and reduced PDGF-D expression . Bayesian hierarchical model analysis has demonstrated a significant relationship between CAD GWAS meta-analysis data and vascular tissue expression data for PDGF-D . The genetic evidence is supported by functional studies showing that PDGF-D influences vascular smooth muscle cell behavior, potentially contributing to the development of atherosclerotic lesions through effects on cell proliferation, migration, and vascular remodeling.
PDGF-D acts as a potent mesangial cell mitogen and contributes significantly to kidney disease progression . In mouse models with adenovirus-mediated overexpression of PDGF-D, animals developed severe mesangial proliferative glomerulopathy characterized by enlarged glomeruli and dramatically increased glomerular cellularity . This pathological response was more severe than that observed with PDGF-B overexpression, while PDGF-C overexpression showed no measurable response . In quantitative terms, mice overexpressing PDGF-B showed a mild increase in glomerular size (4343.5 μm²) and number of cells per glomerular tuft (78), while PDGF-D-expressing mice exhibited more dramatic changes . These findings suggest that PDGF-D signaling represents a potential therapeutic target for mesangial proliferative kidney diseases.
Multiple lines of evidence support PDGF-D's role in cancer progression, with particularly strong data for prostate cancer :
Autocrine signaling: LNCaP prostate cancer cells can auto-activate latent PDGF-D, which induces phosphorylation of the β-PDGF receptor and stimulates cell proliferation in an autocrine manner .
Paracrine effects: LNCaP-PDGF-D-conditioned medium induces migration of prostate fibroblast cell line 1532-FTX, indicating PDGF-D also functions in a paracrine manner to influence stromal cells .
In vivo tumor growth: In a severe combined immunodeficient mouse model, PDGF-D expression accelerates early onset of prostate tumor growth and dramatically enhances prostate carcinoma cell interaction with surrounding stromal cells .
These findings demonstrate PDGF-D's potential oncogenic activity in prostate cancer through both direct effects on cancer cells and modulation of tumor-stromal interactions that support cancer progression.
Several animal models have been developed to study PDGF-D function in vivo:
Transgenic mice expressing PDGF-D in basal epidermal cells: These mice exhibit increased macrophage numbers and elevated interstitial fluid pressure in the dermis, making them valuable for studying PDGF-D's role in skin biology and wound healing .
Adenovirus-mediated PDGF-D delivery models: These models allow for controlled, time-limited expression of PDGF-D in specific tissues and have been particularly useful for studying renal pathology, showing that PDGF-D induces severe mesangial proliferative glomerulopathy .
AAV vector models with tissue-specific promoters: AAV vectors carrying PDGF-D under the control of the RPE-specific VDM2 promoter have been used to study PDGF-D function in retinal tissues .
Xenograft models with PDGF-D-expressing cancer cells: Severe combined immunodeficient mice implanted with PDGF-D-expressing LNCaP cells have helped elucidate PDGF-D's role in prostate cancer progression .
These diverse models allow researchers to investigate PDGF-D function in different physiological and pathological contexts.
Multiple techniques are available for assessing PDGF-D expression and activity:
Gene expression analysis:
Protein detection:
Functional assays:
In vivo assessment:
These complementary approaches provide a comprehensive assessment of PDGF-D expression, processing, and biological activity.
Researchers can manipulate PDGF-D signaling through several approaches:
Genetic manipulation of PDGF-D expression:
Receptor-level interventions:
Ligand-level interventions:
Recombinant PDGF-D protein administration
Anti-PDGF-D neutralizing antibodies
Inhibition of proteases responsible for PDGF-D activation
Downstream signaling manipulation:
Pharmacological inhibitors of key signaling nodes
Genetic manipulation of downstream effectors
When designing interventions, researchers should consider cell type-specific responses, compensatory mechanisms, and potential roles of PDGF receptor heterodimers or crosstalks with other signaling pathways.
While we know PDGF-D requires proteolytic processing for activation, the specific proteases involved in different tissues and disease states remain incompletely characterized . Future research should focus on identifying these proteases and their regulation in various contexts, particularly in cancer and inflammatory conditions. Methodological approaches might include protease inhibitor screens, proteomics analysis of PDGF-D-associated proteins, and targeted gene editing of candidate proteases. Understanding tissue-specific activation mechanisms could reveal new therapeutic targets for modulating PDGF-D activity in a context-dependent manner.
Given PDGF-D's potent effects on mesangial cell proliferation and its role in glomerular pathology , developing targeted therapies for mesangial proliferative diseases represents an important research direction. Future investigations should explore:
Delivery methods for kidney-specific PDGF-D inhibition
Development of antibodies or small molecules that selectively inhibit PDGF-D without affecting other PDGF family members
Identification of downstream mediators unique to PDGF-D signaling in mesangial cells
Combination therapies targeting both PDGF-D and complementary pathways
Platelet-Derived Growth Factor-D (PDGF-D) is a member of the PDGF family, which plays a crucial role in regulating cell growth, proliferation, and angiogenesis. PDGF-D, like other PDGFs, is a potent mitogen for cells of mesenchymal origin, including fibroblasts, smooth muscle cells, and glial cells .
PDGF-D is a dimeric glycoprotein composed of two disulfide-linked subunits. It is one of the five known isoforms of PDGF, which include PDGF-AA, PDGF-BB, PDGF-CC, PDGF-DD, and the heterodimer PDGF-AB . PDGF-D specifically binds to the PDGFRβ receptor, which is a receptor tyrosine kinase (RTK). Upon binding, the receptor dimerizes and undergoes autophosphorylation, activating various downstream signaling pathways such as the PI3K pathway and the STAT3 pathway .
PDGF-D is involved in several critical biological processes:
Recombinant PDGF-D has several applications in medicine and research: