PDGF-CC (Platelet-Derived Growth Factor CC) is a homodimeric growth factor derived from the PDGF family, encoded by the PDGFC gene . It plays critical roles in angiogenesis, tissue development, and stem cell regulation. Structurally, PDGF-CC exists as a latent homodimer requiring proteolytic cleavage to release its bioactive form, which binds selectively to the PDGFR-α receptor tyrosine kinase .
PDGF-CC activates PDGFR-α homodimers and induces α/β heterodimers when PDGFR-β is co-expressed . This dual signaling capacity enables diverse cellular responses.
Receptor | Binding Affinity | Signaling Outcome |
---|---|---|
PDGFR-α | High | Homodimerization, MAPK/PI3K pathway activation |
PDGFR-β | Indirect | Transactivation via α/β heterodimers |
Embryogenesis: Critical for ductal morphogenesis, palatogenesis, and smooth muscle development . PDGF-C knockout mice exhibit cleft palate due to impaired mesenchymal cell migration .
Tissue Morphogenesis: Regulates vascular smooth muscle proliferation and coronary artery development .
Angiogenesis: Stimulates vessel growth in mouse cornea and chick chorioallantoic membrane (CAM) assays .
Stem Cell Dynamics:
Tumorigenesis: Overexpression drives tumor growth, angiogenesis, and fibrosis .
Fibrotic Disorders: Contributes to extracellular matrix remodeling in organs like the liver and lungs .
Wound Healing: Enhances fibroblast proliferation and collagen synthesis .
Cardiovascular Repair: Promotes revascularization of ischemic tissues via EPC activation .
PDGF-CC is a member of the platelet-derived growth factor family discovered in 2000, approximately 20 years after PDGF-A and PDGF-B . Unlike other family members, PDGF-CC primarily binds to PDGFR-α, though it can engage PDGFR-β when co-expressed with PDGFR-α .
Methodological approach: To distinguish PDGF-CC from other family members in research, use specific antibodies that recognize unique epitopes in immunoassays. Western blotting can identify both the full-length latent form and the proteolytically processed active form, which have distinct molecular weights. When designing experiments involving PDGF family members, include appropriate controls to verify specificity of detection methods.
PDGF-CC primarily activates multiple downstream pathways through PDGF receptors, including Ras-MAPK, PI3K/Akt, and PLC-γ signaling cascades . These pathways regulate diverse cellular processes including proliferation, survival, migration, and metabolism.
Methodological approach: To investigate these pathways experimentally, use phospho-specific antibodies to detect activated signaling molecules following PDGF-CC stimulation. Employ pathway-specific inhibitors to validate the contribution of individual signaling branches to observed biological effects. Time-course experiments are essential to capture both immediate and delayed signaling events. For comprehensive analysis, consider phosphoproteomics to identify all phosphorylation events triggered by PDGF-CC stimulation.
PDGF-CC is generally expressed in most human organs and cell types , though expression levels vary significantly between tissues. It plays important roles in various biological processes including development, angiogenesis, tumor growth, tissue remodeling, and stem cell regulation.
Methodological approach: Map PDGF-CC expression using immunohistochemistry on tissue microarrays with validated antibodies. Complement with RNA-seq or qRT-PCR to quantify transcript levels across tissues. Single-cell analyses can reveal cell type-specific expression patterns within heterogeneous tissues. When reporting expression data, normalize to appropriate housekeeping genes and include positive and negative control tissues.
The choice of method depends on the sample type and research question.
Methodological approach: For serum or plasma samples, ELISA provides quantitative measurement of PDGF-CC concentration, as demonstrated in studies of Kawasaki disease where serum PDGF-CC was significantly elevated (3.504 ng/ml) compared to controls (1.237 ng/ml) . For tissue samples, immunohistochemistry provides information on cellular localization, while Western blotting can distinguish between latent and active forms. Ensure proper sample handling to prevent degradation, and include appropriate controls (positive, negative, and recombinant standards) in all assays. Consider potential cross-reactivity with other PDGF family members when selecting antibodies.
Methodological approach: Several strategies can be employed:
Genetic approaches: CRISPR-Cas9 knockout of PDGF-CC, as used in HuCCT1 and QBC939 cholangiocarcinoma cell lines, demonstrated significantly reduced proliferation, migration, invasion, and colony formation in vitro .
RNA interference: siRNA or shRNA targeting PDGF-CC for transient or stable knockdown.
Neutralizing antibodies: Block PDGF-CC activity without affecting expression.
Pharmacological inhibitors: Target PDGF-CC receptors or downstream signaling pathways.
When implementing these approaches, validate modulation at both mRNA (qRT-PCR) and protein (Western blot, ELISA) levels before interpreting downstream effects.
Methodological approach: Rigorous experimental design requires:
Specificity controls: Demonstrate antibody specificity using PDGF-CC knockout samples or blocking peptides.
Functional validation: Confirm that recombinant PDGF-CC activates known downstream pathways.
Dose-response relationships: Establish concentration-dependent effects to ensure biological relevance.
Multiple model systems: Validate findings across different cell lines or animal models to ensure generalizability.
Rescue experiments: Re-introduce PDGF-CC in knockout systems to confirm phenotype specificity.
Comparison with other PDGF family members: Include PDGF-AA, PDGF-BB, and PDGF-DD to assess specificity of observed effects.
PDGF-CC has emerging potential as a biomarker in several conditions:
Methodological approach: For biomarker validation studies:
PDGF-CC plays multiple roles in cancer progression:
Methodological approach: To investigate PDGF-CC in cancer:
Expression analysis: Quantify PDGF-CC in matched tumor and normal tissues using qRT-PCR, Western blotting, and immunohistochemistry.
Functional assays: In cholangiocarcinoma studies, PDGF-CC knockout significantly reduced proliferation, migration, invasion, and colony formation in vitro, while also decreasing tumor growth in xenograft models .
Mechanism investigation: Analyze downstream signaling pathways (PI3K/Akt, Ras/MAPK) activated by PDGF-CC in cancer cells.
Clinical correlation: Assess relationship between PDGF-CC expression and patient outcomes using Kaplan-Meier survival analysis and multivariate regression models.
Therapeutic targeting: Evaluate effects of PDGF-CC inhibition on tumor growth and response to standard therapies.
PDGF-CC is implicated in various inflammatory processes:
Methodological approach: For studying PDGF-CC in inflammation:
In Kawasaki disease, PDGF-CC levels positively correlate with white blood cell counts, percentage of neutrophils, and inflammatory cytokines (IL-2, IL-12p70, TNF-α, IL-1β) .
Design time-course studies to determine whether PDGF-CC elevation precedes, coincides with, or follows inflammatory responses.
Use cell-specific conditional knockout models to determine which cell types contribute to or respond to PDGF-CC during inflammation.
Analyze effects of PDGF-CC on immune cell recruitment, activation, and cytokine production using flow cytometry and multiplex cytokine assays.
Investigate whether PDGF-CC blockade modifies disease course in inflammatory models.
PDGF-CC functions within complex cellular networks:
Methodological approach: To investigate these interactions:
Use proximity ligation assays to detect molecular interactions between PDGF-CC and extracellular matrix components or other growth factors.
Employ 3D culture systems with defined matrix components to study how ECM composition affects PDGF-CC activity.
Analyze synergistic or antagonistic effects when PDGF-CC is combined with other growth factors like VEGF, which has been linked to PDGF-CC in angiogenesis studies.
Utilize decellularized tissue matrices to examine PDGF-CC binding, retention, and release under physiologically relevant conditions.
Apply computational modeling to predict interaction networks based on experimental data.
PDGF-CC requires proteolytic processing for activation:
Methodological approach: To overcome challenges in studying this process:
Develop antibodies specific to latent versus active forms of PDGF-CC to monitor activation status.
Design reporter systems that signal when PDGF-CC is cleaved, allowing real-time monitoring of activation.
Identify and characterize the tissue-specific proteases responsible for PDGF-CC activation in different physiological contexts.
Create mutant PDGF-CC variants resistant to proteolytic activation to determine the functional importance of this regulatory mechanism.
Use protease inhibitor panels to identify which proteases are responsible for PDGF-CC activation in specific tissues or disease states.
Single-cell approaches offer unprecedented resolution:
Methodological approach: To leverage these technologies:
Apply single-cell RNA sequencing to identify cell populations that express PDGF-CC or its receptors within heterogeneous tissues.
Use CyTOF or spectral flow cytometry to simultaneously measure multiple signaling nodes activated by PDGF-CC at single-cell resolution.
Implement spatial transcriptomics to map PDGF-CC signaling networks while preserving tissue architecture information.
Develop single-cell CRISPR screens to identify genes that modify cellular responses to PDGF-CC.
Integrate multiple single-cell data modalities to comprehensively characterize PDGF-CC signaling heterogeneity.
Multiple approaches show promise:
Methodological approach: For therapeutic development:
Neutralizing antibodies against PDGF-CC provide high specificity but may have limited tissue penetration.
Small molecule inhibitors targeting PDGF receptors offer broader coverage but may affect signaling by other PDGF ligands.
Inhibition of proteases required for PDGF-CC activation presents an indirect targeting strategy.
In cholangiocarcinoma research, PDGF-CC inhibition significantly decreased tumor growth in xenograft models, suggesting therapeutic potential .
Design combination strategies that target PDGF-CC signaling alongside standard therapies to overcome resistance mechanisms.
PDGF-CC expression varies significantly between patients:
Methodological approach: For effective stratification:
Develop standardized assays to quantify PDGF-CC in patient samples (tissue, serum) with established reference ranges.
Determine clinically relevant cutoff values that separate patient populations with distinct outcomes or treatment responses.
In cholangiocarcinoma, high PDGF-CC expression correlates with poor survival, providing a rationale for stratifying patients in clinical trials of PDGF-CC inhibitors .
Implement companion diagnostic development alongside therapeutic agents targeting PDGF-CC pathways.
Design adaptive trial protocols that adjust treatment based on changes in PDGF-CC levels during therapy.
PDGF-CC has emerging roles in stem cell biology:
Methodological approach: To investigate these functions:
Study effects of recombinant PDGF-CC on proliferation, self-renewal, and differentiation of various stem cell populations.
Compare stem cell behaviors in wild-type versus PDGF-CC knockout models during development and tissue regeneration.
Analyze PDGF-CC expression patterns during embryonic development and adult tissue repair processes .
Investigate whether PDGF-CC supplementation can enhance stem cell therapy outcomes in preclinical models.
Determine cell type-specific responses to PDGF-CC among different progenitor populations using lineage tracing and single-cell analysis.
PDGF-CC activates multiple signaling pathways critical for cell function:
Receptor binding: Primarily binds PDGFR-α but can engage PDGFR-β when co-expressed with PDGFR-α
Major activated pathways: Ras-MAPK, PI3K/Akt, and PLC-γ signaling
Biological responses: Regulates cell proliferation, survival, migration, angiogenesis, and metabolism
In cancer: Promotes tumor cell growth, invasion, metastasis, and therapy resistance
In inflammation: Correlates with inflammatory markers and cytokine expression
Platelet-Derived Growth Factor-CC (PDGF-CC) is a member of the PDGF family, which consists of proteins derived from four genes: PDGF-A, PDGF-B, PDGF-C, and PDGF-D. These proteins form four disulfide-linked homodimers (PDGF-AA, PDGF-BB, PDGF-CC, and PDGF-DD) and one heterodimer (PDGF-AB) . PDGF-CC, like other PDGFs, plays a crucial role in regulating cell growth and division, particularly in the context of blood vessel formation and the proliferation of mesenchymal cells .
PDGF-CC is a dimeric glycoprotein composed of two C subunits. It is a potent mitogen for cells of mesenchymal origin, including fibroblasts, smooth muscle cells, and glial cells . The PDGF family members, including PDGF-CC, function as secreted, disulfide-linked homodimers. PDGF-CC specifically interacts with the PDGFRα (PDGFRA) receptor, which is a receptor tyrosine kinase (RTK) .
The receptor for PDGF-CC, PDGFRα, binds to the ligand and undergoes dimerization and autophosphorylation. This activation triggers downstream signaling pathways, such as the PI3K pathway and the STAT3 pathway, which are involved in cell proliferation, migration, and survival . PDGF-CC is particularly significant in the context of blood vessel formation, also known as angiogenesis, and the growth of blood vessels from pre-existing blood vessel tissue .