The IFI30 antibody targets the IFI30 protein (UniProt: P13284), also known as Gamma-Interferon-Inducible Lysosomal Thiol Reductase (GILT). Key characteristics include:
Gliomas: High IFI30 expression correlates with glioblastoma (GBM) malignancy, mesenchymal subtype, and poor prognosis. It independently predicts 1-, 3-, and 5-year survival rates .
Zebrafish Models: Ifi30 knockdown disrupts caudal vein plexus (CVP) formation and intersegmental vessel sprouting. Phenotypes include vessel rupture and pericardial edema, indicating its necessity for vascular development .
Western Blot: Detects IFI30 in human colon tissue at dilutions of 1:500–1:5000 .
Immunohistochemistry (IHC): Shows elevated IFI30 in high-grade gliomas (p < 0.01) .
ELISA: Validated for quantitative analysis of IFI30 in immune cell lysates .
Biomarker Potential: IFI30 levels stratify glioma subtypes and predict chemotherapy response .
Immune Modulation: Linked to antigen presentation defects in melanoma and breast cancer, where low IFI30 correlates with poor survival .
Therapeutic Target: Preclinical studies suggest targeting IFI30 could enhance chemotherapy efficacy or reprogram immunosuppressive microenvironments .
IFI30, also known as gamma-interferon-inducible lysosomal thiol reductase (GILT), is a protein that plays critical roles in various biological processes. In humans, the canonical protein consists of 250 amino acid residues with a molecular mass of approximately 28 kDa . It localizes primarily to lysosomes and is also secreted . IFI30 is important in research because it:
The protein has gained significant research interest due to its aberrant expression in several cancer types, including diffuse large B-cell lymphoma (DLBCL), melanoma, breast cancer, and glioma .
IFI30 is a member of the GILT protein family and is characterized by several important biochemical and structural features:
Molecular weight: Calculated at 29 kDa based on 261 amino acids; observed at 29 kDa in experimental conditions
Post-translational modifications: Undergoes N-glycosylation and protein cleavage
Subcellular localization: Primarily found in lysosomes and is also secreted into the extracellular space
Expression pattern: Widely expressed across many human tissue types
Orthologs: Present in mouse, rat, bovine, frog, chimpanzee, and chicken species
IFI30 antibodies are versatile tools in molecular and cellular research, with applications including:
These applications enable researchers to study IFI30 expression, localization, and function in various experimental models .
Optimization of antibody dilution is critical for obtaining reliable and reproducible results in IFI30 research. The following methodological approach is recommended:
For Western Blot applications:
Begin with a moderate dilution (1:5000) within the recommended range (1:2000-1:12000)
Perform a titration experiment with serial dilutions if signal strength is suboptimal
Consider cell/tissue type specificity, as observed in COLO 320 cells
Validate specificity with appropriate positive and negative controls
For Immunohistochemistry:
For Immunofluorescence/ICC:
For all applications, titration experiments with different antibody concentrations are essential to determine optimal conditions for your specific experimental system.
The detection of IFI30 requires specific sample preparation methods depending on the tissue type and analytical technique:
Use standard lysis buffers containing protease inhibitors
Fixation: Standard formalin fixation and paraffin embedding
Antigen retrieval: Primary recommendation is TE buffer pH 9.0
Alternative method: Citrate buffer pH 6.0 if primary method yields suboptimal results
Fixation: Standard 4% paraformaldehyde
Permeabilization: 0.1-0.5% Triton X-100
Blocking: 1-5% BSA or serum appropriate to secondary antibody species
Each preparation method should be optimized for the specific research question and tissue type under investigation.
IFI30 has emerged as a significant factor in cancer biology, particularly in relation to tumor progression and therapy resistance. IFI30 antibodies can be employed in several sophisticated research applications:
Analysis of epithelial-mesenchymal transition (EMT) processes:
IFI30 promotes the EMT-like phenotype in glioma cells by activating the EGFR/AKT/GSK3β/β-catenin pathway
Use IFI30 antibodies in co-immunoprecipitation experiments to identify protein-protein interactions within this pathway
Combine with antibodies against EMT markers (E-cadherin, vimentin, Slug) for multiplexed immunofluorescence to visualize transition states
Investigation of chemoresistance mechanisms:
IFI30 regulates chemoresistance to temozolomide (TMZ) in glioma cells
Expression levels increase in response to TMZ treatment in a dose-dependent manner
Design experiments comparing IFI30 expression levels in parental versus TMZ-resistant cell lines using quantitative Western blot analysis
Correlate with patient response data using tissue microarrays and IHC
Translational research applications:
Prognostic marker development: High IFI30 expression correlates with poor patient outcomes in glioblastoma
Target identification: IFI30 represents a potential therapeutic target for TMZ-resistant glioma
Patient stratification: Consider IFI30 expression levels when designing clinical trials for glioma therapies
To determine the functional significance of IFI30, researchers can implement several sophisticated experimental strategies:
Gene modulation techniques:
Knockdown approaches: shRNA targeting IFI30 has been effective in GSC464 cells, resulting in decreased tumor volume and weight in xenograft models
Overexpression studies: Transfection of IFI30 expression constructs enhances mesenchymal characteristics in glioma cell lines
CRISPR/Cas9 genome editing for complete knockout studies
Functional assays for cancer-related phenotypes:
Cell viability: CCK-8 assays reveal decreased viability after IFI30 knockdown
Colony formation: Quantification of colony-forming capacity after IFI30 modulation
Cell migration: Wound healing and transwell assays demonstrate reduced migration following IFI30 silencing
Limiting dilution analysis: Evaluates self-renewal capacity of cancer stem cells with altered IFI30 expression
Pathway analysis and signaling studies:
When working with IFI30 antibodies, researchers frequently encounter several technical challenges. Here are methodological approaches to address these issues:
Specificity validation concerns:
Conduct parallel experiments with multiple antibody clones targeting different epitopes
Include appropriate controls: positive control (tissue/cells known to express IFI30), negative control (IFI30 knockout/knockdown samples)
Verify specificity with IP-Western blot confirmation
Consider pre-adsorption tests with immunizing peptide when available
Background signal in immunohistochemistry:
Optimize blocking conditions: Increase blocking time or concentration (5% BSA or normal serum)
Adjust antibody concentration: Start with higher dilutions (1:8000) and titrate as needed
Modify antigen retrieval: Switch between recommended TE buffer pH 9.0 and alternative citrate buffer pH 6.0
Reduce secondary antibody concentration or use more specific detection systems
Variable detection across different sample types:
Tissue-specific optimization may be required as IFI30 is widely expressed in many tissues
Adjust protein extraction protocols based on tissue type (e.g., brain tissue vs. cultured cells)
Consider post-translational modifications: IFI30 undergoes N-glycosylation and cleavage that may affect detection
Interpreting conflicting IFI30 expression data requires careful consideration of multiple factors:
Cellular context considerations:
Methodological factors affecting expression analysis:
RNA vs. protein detection: IFI30 mRNA levels (qPCR, RNA-seq) may not directly correlate with protein expression (Western blot, IHC)
Antibody selection: Different antibody clones may have varying affinities for different IFI30 isoforms or post-translationally modified forms
Sample preparation: Fixation methods for IHC can affect epitope accessibility
Experimental intervention effects:
Treatment-induced changes: TMZ treatment upregulates IFI30 expression in glioma cells
Temporal dynamics: Consider time-course experiments as IFI30 expression may change during disease progression
Microenvironmental influences: IFI30 is interferon-inducible, so inflammatory conditions in the tumor microenvironment may influence expression levels
When faced with conflicting data, a comprehensive approach involving multiple detection methods, careful consideration of experimental conditions, and validation across independent sample sets is recommended.
Based on current knowledge, several promising research directions for IFI30 antibodies are emerging:
Biomarker development for cancer prognosis:
Therapeutic resistance mechanisms:
IFI30 upregulation occurs in response to temozolomide treatment
Antibody-based detection of IFI30 could identify patients likely to develop resistance
Targeting the EGFR/AKT/GSK3β/β-catenin pathway influenced by IFI30 may overcome resistance
Combination therapy approaches based on IFI30 expression patterns
Immunotherapy connections:
Given IFI30's role in MHC class II-restricted antigen processing
Investigation of correlations between IFI30 expression and immunotherapy response
Potential for enhancing immune recognition of tumors by modulating IFI30 function
Development of novel immunotherapeutic approaches targeting IFI30-related pathways
Advanced multiparametric approaches incorporating IFI30 antibodies offer significant potential for cancer research:
Integrated molecular classification systems:
Spatial analysis of tumor heterogeneity:
Multiplex immunofluorescence incorporating IFI30 antibodies with EMT markers
Spatial transcriptomics combined with IFI30 protein detection
Analysis of IFI30 expression in relation to tumor microenvironment components
Correlation with invasive tumor fronts vs. tumor core regions
Therapeutic decision support:
These emerging applications highlight the importance of incorporating IFI30 antibodies into comprehensive research strategies aimed at improving cancer classification, treatment selection, and patient outcomes.