PAFAH1B3 is overexpressed in multiple cancers and correlates with poor prognosis. Key findings include:
Immune Regulation: PAFAH1B3 correlates with tumor mutational burden (TMB), microsatellite instability (MSI), and immune cell infiltration in cancers like LIHC and BRCA .
Therapeutic Target: Inhibition upregulates tumor-suppressing lipids, sensitizing cells to immunotherapy .
PAFAH1B3 silencing increases sub-G1 phase cells (apoptosis) and induces G0/G1 arrest in HCC .
Co-expressed genes (e.g., CDK1, PLK1) drive mitotic transitions and tumor growth .
Modulates glycolysis and lipid metabolism pathways, critical for cancer cell survival .
Inhibition reduces ATP production and fatty acid synthesis in HCC .
High PAFAH1B3 expression correlates with immunosuppressive markers (e.g., PD-L1) and reduced CD8+ T-cell infiltration .
PAFAH1B3 is a promising target for cancer therapy:
PAFAH1B3 is the alpha1 catalytic subunit of the cytosolic type I platelet-activating factor acetylhydrolase (PAF-AH I), a heterotetrameric enzyme that catalyzes the hydrolysis of the acetyl group at the sn-2 position of PAF and related molecules . It functions as either an alpha1/alpha1 homodimer or an alpha1/alpha2 heterodimer with PAFAH1B2, with different compositions affecting enzyme activity and substrate specificity .
Beyond its enzymatic role, PAFAH1B3 participates in multiple signaling pathways including:
Functionally, PAFAH1B3 plays critical roles in:
When investigating PAFAH1B3 expression, researchers typically employ multiple complementary techniques:
Protein-level detection:
Immunohistochemistry (IHC) - The most common method for clinical specimens, used to examine expression patterns in HSCC tissues and other cancers
Western blotting - For quantitative protein expression analysis in cell lines and tissue samples
Proteomic analysis - Using mass spectrometry for unbiased protein quantification
mRNA-level analysis:
qRT-PCR - For quantitative mRNA expression measurement
RNA-seq - For comprehensive transcriptomic profiling, often used in public database analyses like TCGA
In situ hybridization - For spatial expression analysis in tissues
TCGA and other public databases provide valuable resources for examining PAFAH1B3 expression across cancer types and correlating with clinical parameters . When designing studies, researchers should consider both transcriptomic and proteomic approaches, as post-transcriptional regulation may result in discrepancies between mRNA and protein levels.
PAFAH1B3 demonstrates widespread upregulation across numerous cancer types, positioning it among the 50 most commonly upregulated metabolic enzymes across more than 1,000 primary human tumors spanning 19 cancer types .
Cancer types with confirmed PAFAH1B3 overexpression:
The evidence for this upregulation comes from multiple methodologies, including TCGA database analysis, proteomic analysis through the UALCAN database, and direct immunohistochemical evaluation of clinical specimens . For example, in HSCC studies, PAFAH1B3 was significantly overexpressed in tumor tissues compared to paired adjacent non-tumor samples (p<0.0001) .
PAFAH1B3 functionally influences multiple aspects of cancer cell behavior, as demonstrated through loss-of-function studies in various cancer models:
Cell proliferation:
Knockdown of PAFAH1B3 significantly inhibits cell proliferation in liver hepatocellular carcinoma (LIHC) cell lines
Similar antiproliferative effects observed in HSCC FaDu cells
Cell migration and invasion:
PAFAH1B3 silencing reduces migration and invasion capabilities in LIHC cancer models
Comparable inhibition of these metastatic properties confirmed in HSCC models
Mechanistic basis:
PAFAH1B3 maintains tumor cell aggressiveness through:
Possible involvement in PAF-related signaling cascades affecting cell growth and metastasis
Participation in Wnt signaling pathways, known regulators of cell proliferation and stemness
In experimental designs, researchers should consider both short-term proliferation assays (MTT, BrdU incorporation) and longer-term colony formation assays, alongside migration/invasion assays (wound healing, transwell) to comprehensively evaluate the impact of PAFAH1B3 manipulation.
PAFAH1B3 demonstrates significant associations with immune parameters in the tumor microenvironment, suggesting an immunomodulatory role:
Immune cell infiltration:
PAFAH1B3 expression positively correlates with immune cell infiltration across cancer types
Different expression patterns observed in various immune subtypes of cancers (e.g., distinct patterns in different immune subtypes of LIHC)
Molecular correlations:
Pathway involvement:
Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis confirms that PAFAH1B3 is primarily involved in immune regulation pathways .
When investigating PAFAH1B3's immune interactions, researchers should consider implementing:
Immune cell deconvolution analyses of bulk RNA-seq data
Flow cytometry to quantify immune populations in experimental models
Single-cell RNA-seq to resolve specific immune cell type associations
Co-culture systems to study direct interactions between PAFAH1B3-expressing cancer cells and immune components
PAFAH1B3 demonstrates significant prognostic value across multiple cancer types, with high expression consistently associated with worse clinical outcomes:
Disease-specific survival (DSS):
Negative correlation with DSS in:
Progression-free interval (PFI):
Elevated PAFAH1B3 associated with shorter PFI in multiple cancers, most notably LIHC and NSCLC .
These survival correlations have been established through rigorous statistical analyses of large patient cohorts, including multivariate analyses confirming PAFAH1B3 as an independent prognostic factor in several cancer types .
PAFAH1B3 has demonstrated potential as an independent prognostic biomarker, particularly in liver hepatocellular carcinoma (LIHC) and hypopharyngeal squamous cell carcinoma (HSCC):
Liver hepatocellular carcinoma (LIHC):
Hypopharyngeal squamous cell carcinoma (HSCC):
High PAFAH1B3 expression independently associated with poor prognosis
Significant correlation with lymph node metastasis and advanced clinical stage
Methodological considerations for biomarker validation:
When evaluating PAFAH1B3 as a prognostic biomarker, researchers should:
Use Cox proportional hazards models for multivariate analyses
Include established clinicopathological parameters as covariates
Apply Kaplan-Meier survival analysis with log-rank tests for visualization
Consider ROC curve analysis to establish optimal expression thresholds
Validate findings in independent patient cohorts
PAFAH1B3's prognostic value appears most robust in hepatocellular and certain lung cancer subtypes, suggesting prioritization of these cancer types for further biomarker development efforts .
When designing PAFAH1B3 loss-of-function studies, researchers have successfully employed several RNA interference approaches:
siRNA-mediated knockdown:
Advantages: Rapid implementation, high efficacy, commercially available reagents
Limitations: Transient effect, potential off-target effects, variable transfection efficiency
shRNA-mediated knockdown:
Useful for stable suppression of PAFAH1B3
Advantages: Long-term studies possible, selection for positive cells
Considerations: Requires viral delivery systems, longer implementation timeline
CRISPR/Cas9 gene editing:
For complete knockout studies
Advantages: Complete elimination of protein expression, isogenic control creation
Considerations: Potential compensation by PAFAH1B2, phenotype may differ from knockdown
Methodological recommendations:
Validate knockdown efficiency at both mRNA level (qRT-PCR) and protein level (Western blot)
Include multiple independent siRNA/shRNA constructs to confirm specificity
Consider rescue experiments to confirm phenotype specificity
For cancer studies, both LIHC and HSCC cell line models have proven effective
When interpreting results, researchers should remain aware that complete knockout may have different effects than partial knockdown, potentially due to the compensatory role of PAFAH1B2 or other metabolic adaptations.
PAFAH1B3 expression correlates with drug sensitivity profiles across cancer types, presenting opportunities for therapeutic research:
Database-driven approaches:
The Genomics of Drug Sensitivity in Cancer (GDSC) database analysis reveals correlations between PAFAH1B3 expression and drug response
Cancer Therapeutics Response Portal (CTRP) provides additional drug sensitivity data
These resources allow in silico identification of potential drug-PAFAH1B3 interactions
Experimental methods:
Cell line panels: Testing drug sensitivity across lines with varying PAFAH1B3 expression
Knockdown/overexpression studies: Evaluating how PAFAH1B3 manipulation alters drug response
Combination studies: Testing PAFAH1B3 inhibitors with established therapeutics
Special considerations:
PAFAH1B3 has been identified as a target for combination therapy with tyrosine kinase inhibitors (TKIs) in certain leukemias
When studying drug interactions, both synergistic and antagonistic effects should be evaluated using combination index calculation methods
Patient-derived models may better recapitulate the heterogeneity of drug responses
For optimal experimental design, researchers should:
Include multiple concentrations to generate complete dose-response curves
Examine temporal dynamics of response
Consider both cytostatic (growth inhibition) and cytotoxic (cell death) endpoints
Validate findings across multiple cell line models
Genetic alterations in PAFAH1B3 can significantly impact its expression, function, and prognostic associations:
Types of observed alterations:
Mutations in the PAFAH1B3 gene
Copy number variations
Translocations (a translocation between PAFAH1B3 on chromosome 19 and CDC-like kinase 2 gene on chromosome 1 has been observed)
Functional consequences:
Genetic alterations in PAFAH1B3 affect its expression levels and prognostic ability
Some alterations are associated with cognitive disability, ataxia, and brain atrophy
In cancer contexts, alterations may enhance or diminish PAFAH1B3's oncogenic properties
Research methodologies:
Mutation analysis: Using TCGA or COSMIC databases to identify recurring mutations
Structure-function studies: Site-directed mutagenesis to examine how specific alterations affect enzyme activity
Copy number analysis: Correlating CNV with expression and phenotypic changes
Clinical correlation: Associating specific alterations with treatment response or outcomes
When investigating PAFAH1B3 alterations, researchers should consider both somatic mutations in cancer contexts and germline variants that may predispose to disease or affect development.
PAFAH1B3 influences cancer biology through several mechanistic pathways:
Lipid metabolism modulation:
PAFAH1B3 maintains tumor cell aggressiveness via regulating tumor-suppressing lipids
As a metabolic enzyme, it impacts lipid homeostasis critical for cancer cell membranes and signaling
Signaling pathway involvement:
PAF signaling pathways: Affecting inflammation and potentially immune response in tumor microenvironment
Wnt pathways: Critical for cell proliferation, stemness, and metastasis
Immune modulation:
Methodological approaches to mechanism studies:
Lipidomics: To identify specific lipid species altered by PAFAH1B3 manipulation
Pathway reporter assays: To quantify effects on Wnt, PAF, and other signaling pathways
Protein-protein interaction studies: Co-IP or proximity labeling to identify binding partners
Transcriptomics: RNA-seq after PAFAH1B3 manipulation to identify downstream effectors
Understanding these mechanisms provides potential opportunities for therapeutic intervention and biomarker development across multiple cancer types.
Given PAFAH1B3's role in cancer, several therapeutic approaches are being investigated:
Direct enzymatic inhibition:
Small molecule inhibitors targeting PAFAH1B3's catalytic activity
Structure-based drug design leveraging known protein structure
High-throughput screening to identify novel inhibitors
Gene expression modulation:
siRNA/shRNA-based approaches, potentially deliverable through nanoparticles
Antisense oligonucleotides to reduce PAFAH1B3 expression
PROTAC or molecular glue approaches for protein degradation
Combination strategies:
PAFAH1B3 has been identified as a target for combination therapy with tyrosine kinase inhibitors (TKIs) in BCR-ABL1+ BCP-ALL
Synergistic potential with immune checkpoint inhibitors, given PAFAH1B3's association with immune parameters
Combination with conventional chemotherapies based on drug sensitivity correlations
Target validation considerations:
Cell-type specificity of dependence on PAFAH1B3
Potential compensation by PAFAH1B2 or other pathways
Therapeutic window between cancer and normal tissues
Biomarkers for patient selection (expression level, genetic alterations)
When designing therapeutic studies, researchers should incorporate both pharmacological inhibitors and genetic knockdown approaches to distinguish between catalytic activity-dependent and scaffold function-dependent effects.
PAFAH1B3 research shows promise for several clinical applications:
Prognostic biomarker development:
Particularly in liver hepatocellular carcinoma (LIHC), non-small cell lung cancer (NSCLC), and hypopharyngeal squamous cell carcinoma (HSCC)
Multi-cancer prognostic panels incorporating PAFAH1B3 expression
Integration with other molecular markers for improved risk stratification
Patient stratification:
Identifying high-risk patients who may benefit from more aggressive treatment
Selection of patients for PAFAH1B3-targeted therapies
Potential role in treatment algorithm development for personalized medicine
Therapeutic targets:
Direct PAFAH1B3 inhibition in cancers with strong dependence
Combination approaches, especially with:
Implementation considerations:
Standardization of PAFAH1B3 assessment methods for clinical use
Prospective validation in clinical trials
Development of companion diagnostics alongside therapeutic approaches
Regulatory considerations for biomarker approval
The strongest evidence supports prioritizing liver cancer (LIHC) and lung adenocarcinoma (LUAD) for initial clinical applications, given the robust prognostic associations and functional dependencies demonstrated in these cancer types .
The PAFAH1B3 gene is located on chromosome 19 and encodes a protein that is part of the platelet-activating factor acetylhydrolase isoform 1B complex. This complex consists of three subunits: the catalytic beta (PAFAH1B2) and gamma (PAFAH1B3) subunits, and the regulatory alpha (PAFAH1B1) subunit . The PAFAH1B3 protein itself is approximately 29 kDa in size .
PAFAH1B3 catalyzes the removal of an acetyl group from the glycerol backbone of PAF, converting it into an inactive form known as lyso-PAF . This reaction is essential for regulating the biological activity of PAF and preventing excessive inflammatory responses. The enzyme is involved in various physiological processes, including brain development, spermatogenesis, and the modulation of immune responses .
Mutations or dysregulation of the PAFAH1B3 gene have been associated with several disorders. For instance, a translocation involving this gene and the CDC-like kinase 2 gene on chromosome 1 has been linked to cognitive disabilities, ataxia, and brain atrophy . Additionally, recent studies have indicated that PAFAH1B3 plays a role in cancer progression, particularly in lung adenocarcinoma, where its overexpression is correlated with poor prognosis and increased tumor invasiveness .
Given its involvement in critical biological processes and disease states, PAFAH1B3 is a target of interest for therapeutic interventions. Research is ongoing to better understand its function and regulation, as well as to develop inhibitors that could potentially be used in treating conditions like cancer and inflammatory diseases .