CNPY4 (Protein canopy homolog 4) is a member of the canopy protein family with regulatory functions in multiple signaling pathways. Research indicates that CNPY4 plays critical roles in:
Regulation of cell surface expression of TLR4 (Toll-like receptor 4), indicating involvement in innate immune signaling
Inhibition of the Hedgehog (HH) signaling pathway by modulating membrane sterol composition
Potential regulation of tumor immune microenvironments, particularly in gliomas
The protein contains a SAPLIP (Saposin-like protein) domain that can interact with membrane lipids, suggesting its function in lipid trafficking or extraction for signaling regulation .
CNPY4 is a full-length human protein spanning amino acids 22-248. For research purposes, recombinant CNPY4 can be expressed in Escherichia coli with >85% purity . The protein structure includes:
A saposin-like domain capable of lipid interactions
Multiple cysteine residues forming disulfide bonds
A sequence that allows for proper folding and biological activity
Researchers can obtain the recombinant protein for various applications including SDS-PAGE analysis and mass spectrometry studies .
To analyze CNPY4 expression patterns, researchers commonly employ:
RNA-sequencing and transcriptomic analysis using databases like TCGA
Quantitative PCR for mRNA expression measurement
Western blotting for protein-level detection
Immunohistochemistry for tissue localization studies
Public database mining tools like GEPIA to compare expression across normal and disease tissues
Standardization of detection methods is crucial, as CNPY4 may have tissue-specific expression patterns with meaningful biological implications.
CNPY4 has emerged as a potential prognostic biomarker in gliomas. Analysis of TCGA data reveals:
High CNPY4 expression correlates with poorer prognosis in glioma patients
Patients with low CNPY4 expression demonstrate significantly longer survival times compared to high-expression groups (p < 0.05)
Multivariate analysis confirms that downregulation of CNPY4 expression represents an independent and favorable prognostic factor
These findings suggest CNPY4 expression analysis could provide valuable information for prognostic assessment in clinical settings.
Univariate logistic regression analysis of TCGA data demonstrates that CNPY4 expression correlates with multiple clinicopathological factors:
Tumor grade: Higher expression in higher-grade gliomas
Patient age: Association with age at diagnosis
IDH status: Correlation with IDH mutation status
1p/19q codeletion: Association with this important molecular marker
These correlations suggest CNPY4 may be integrated into molecular classification schemes for gliomas and potentially inform treatment decisions.
CNPY4 expression significantly influences immune cell infiltration patterns in gliomas:
In glioblastoma (GBM), CNPY4 expression positively correlates with dendritic cell infiltration (Partial Cor = 0.28)
In low-grade gliomas (LGG), CNPY4 expression positively correlates with multiple immune cell types:
CIBERSORT analysis further revealed that the proportions of resting NK cells were significantly higher in high CNPY4 expression groups (p = 0.018), while activated NK cells (p = 0.008) and M2 macrophages (p = 0.034) were reduced compared to low expression groups .
CNPY4 functions as a negative regulator of the Hedgehog (HH) pathway through a novel mechanism involving membrane composition modulation:
Knockdown of CNPY4 results in elevated basal activation of the HH transcriptional program
CNPY4 silencing potentiates signaling responses to various HH pathway agonists, including:
Experiments indicate CNPY4 operates in parallel to or downstream of PTCH1 but upstream of SUFU, suggesting its primary effect occurs at the level of Smoothened (SMO) regulation .
A key insight into CNPY4 function involves its ability to modulate membrane sterol accessibility:
Cells with CNPY4 knockdown display significantly elevated levels of accessible membrane sterols compared to control cells
MEFs derived from CNPY4 null animal embryonic limb buds show increased accessible sterol levels in basal states
This effect appears to involve cholesterol, which has been proposed as an endogenous ligand for SMO activation
This mechanism suggests CNPY4 acts as a fine-tuner of Hedgehog signaling by controlling membrane composition, particularly affecting sterol availability for pathway activation.
Several key epistasis experiments have clarified CNPY4's position in the HH signaling cascade:
In PTCH1-/- MEFs (constitutively active HH pathway), CNPY4 knockdown further activates the HH transcriptional program, suggesting CNPY4 acts parallel to or downstream of PTCH1
In SUFU-/- MEFs (also constitutively active), CNPY4 knockdown produces more modest increases in Gli1 expression than in PTCH1-/- cells
In SMO-/- MEFs, SAG or SHH stimulation fails to elicit hyperactive HH signaling after CNPY4 knockdown, indicating that CNPY4, like PTCH1, modulates HH activity through SMO
These findings place CNPY4 action primarily at the level of SMO regulation in the HH pathway.
Researchers can utilize several approaches to manipulate CNPY4 expression:
siRNA knockdown: Transient CNPY4 silencing using siRNA in cell culture models has successfully demonstrated functional impacts on signaling pathways
CRISPR-Cas9 knockout: Complete deletion of CNPY4 has been achieved in mouse embryonic fibroblasts (MEFs) to study developmental and signaling effects
Overexpression systems: Transfection with CNPY4 expression vectors allows for gain-of-function studies
When designing knockdown experiments, researchers should verify efficiency through both mRNA (qPCR) and protein level (Western blot) analyses to ensure complete silencing.
Luciferase reporter assays have proven effective for measuring CNPY4's impact on signaling pathways:
Gli-luciferase reporters can quantify Hedgehog pathway activation following CNPY4 manipulation
These assays can be performed in NIH3T3 cells, which are highly responsive to Hedgehog pathway modulation
Reporter systems allow for comparative analysis of pathway activation under different conditions, including:
Transfection efficiency should be normalized with a constitutive reporter (e.g., Renilla luciferase) for accurate quantification.
To analyze CNPY4's impact on membrane sterol accessibility, researchers can employ:
Modified Perfringolysin O (PFO) probes*: These fluorescently-tagged bacterial toxin derivatives specifically bind accessible sterols in plasma membranes of intact cells
Quantitative imaging analysis: Following PFO* staining, fluorescence microscopy with quantitative image analysis can measure relative sterol accessibility between experimental conditions
Lipidomic profiling: Mass spectrometry-based lipidomics can provide comprehensive analysis of membrane lipid composition changes following CNPY4 manipulation
These techniques provide complementary approaches to understanding CNPY4's mechanistic impact on membrane composition.
CNPY4 significantly impacts immune cell distribution in glioma microenvironments:
CIBERSORT analysis of TCGA data reveals distinct immune cell proportion differences between high and low CNPY4 expression groups:
TIMER analysis demonstrated positive correlations between CNPY4 expression and immune cell infiltration:
These findings suggest CNPY4 may influence antitumor immune responses through modulation of immune cell recruitment and activation states.
Gene Set Enrichment Analysis (GSEA) of TCGA data identified several immune-related pathways significantly associated with high CNPY4 expression:
Immunoregulatory interactions between lymphoid and non-lymphoid cells
Intestinal immune network for IgA production
Autoimmune thyroid disease pathways
Primary immunodeficiencies
These enrichment patterns suggest CNPY4 may influence diverse aspects of immune regulation beyond direct cellular interactions, potentially affecting response to immunotherapies.
CNPY4 has been identified as a regulator of TLR4 (Toll-like receptor 4) cell surface expression . While the detailed mechanism requires further investigation, current understanding suggests:
CNPY4 may influence trafficking of TLR4 to the cell surface, similar to its reported function with other membrane proteins
This regulation could impact innate immune responses to pathogen-associated molecular patterns (PAMPs)
The relationship between CNPY4's lipid-modulating functions and TLR4 regulation represents an interesting area for future research
Researchers investigating this relationship should consider experimental designs that monitor both membrane composition and receptor trafficking dynamics.
CNPY4 is involved in the positive regulation of protein localization to the plasma membrane . It is predicted to enable signaling receptor binding activity, which is essential for various cellular processes . The protein is located in the extracellular region and plays a significant role in the immune response by regulating TLR4 expression .
The human recombinant CNPY4 protein is produced in E. coli as a single, non-glycosylated polypeptide chain . It contains 252 amino acids and is fused to a 25 amino acid His-tag at the N-terminus . The protein is purified using proprietary chromatographic techniques and is available in a sterile filtered colorless solution .