The ITIH3 antibody is a specialized immunological tool targeting Inter-alpha-trypsin inhibitor heavy chain H3 (ITIH3), a glycoprotein involved in extracellular matrix stabilization and inflammatory processes. ITIH3 forms part of the inter-alpha-trypsin inhibitor (ITI) family, which regulates protease activity and hyaluronan metabolism . Antibodies against ITIH3 are critical for research and clinical applications, including biomarker detection, disease monitoring, and mechanistic studies in conditions like myasthenia gravis (MG) and cancer .
ITIH3 antibodies are central to identifying serum biomarkers for MG, an autoimmune neuromuscular disorder. Recent studies demonstrate:
Correlation with Disease Activity: Serum ITIH3 levels, measured via ELISA, correlate strongly with MG disease severity scores (e.g., MG-ADL, QMG) .
Localization at Neuromuscular Junctions: Immunostaining reveals ITIH3 localization at neuromuscular endplates in MG patients, absent in controls, supporting its role in disease pathophysiology .
Treatment Response Prediction: Subgroup analyses show ITIH3 levels predict therapeutic responses, offering a tool for personalized treatment .
ITIH3 antibodies enable investigations into tumor biology and chemotherapy resistance:
Ovarian Cancer: Reduced ITIH3 expression correlates with cisplatin resistance via dysregulation of the Bcl-2 anti-apoptotic pathway. Immunohistochemistry (IHC) confirms ITIH3 as a prognostic marker .
Gastric Cancer: Elevated plasma ITIH3 levels (detected via Western blot) show 96% sensitivity for early-stage detection, making it a promising diagnostic biomarker .
Interaction Partners: Immunoprecipitation coupled with proteomics identified ITIH3-binding proteins (e.g., desmin, plectin) critical for neuromuscular transmission .
Structural Relevance: ITIH3’s localization at endplates provides a structural basis for its serological detection in MG .
Ovarian Cancer:
Gastric Cancer:
Diagnostic Utility: ITIH3 ELISA kits enable non-invasive monitoring of MG progression and treatment efficacy .
Therapeutic Targeting: ITIH3-interacting proteins (e.g., desmin) offer novel targets for modulating neuromuscular function or apoptosis pathways .
ITIH3 is a heavy chain subunit of the alpha trypsin inhibitor complex precursor that plays an essential role in stabilizing the extracellular matrix by preventing hyaluronic acid depolymerization . The protein is encoded by the ITIH3 gene and functions as part of the inter-alpha-trypsin inhibitor (ITI) family. These proteins are known to be involved in various physiological processes, including inflammation, wound healing, and maintenance of tissue integrity.
Research has demonstrated that ITIH3 may serve an important role in tumor progression . Studies have shown that ITIH family genes are markedly downregulated in various human solid tumors, including colon, breast, and lung cancer, suggesting their potential role as tumor suppressor genes . This makes ITIH3 a protein of significant interest in cancer research, particularly in understanding tumor development mechanisms.
Detection of ITIH3 in biological samples typically employs several complementary techniques:
Western Blotting: This technique remains the gold standard for ITIH3 protein expression analysis. Researchers commonly use anti-ITIH3 antibodies with appropriate loading controls such as GAPDH or Actin. For example, studies investigating ITIH3 in ovarian cancer cell lines used western blotting to assess relative protein expression levels in SKOV3, A2780, OVCAR3, and CAOV3 cells .
Immunohistochemistry (IHC): For tissue samples, IHC is frequently employed using specialized antibodies such as goat-anti-ITI-H3 (L-15) antibody (1:200; cat. no. sc-33949; Santa Cruz Biotechnology, Inc.) . The protocol typically involves:
Fixing tissue sections with 4% paraformaldehyde at room temperature for 48 hours
Embedding in paraffin and slicing 4 μm sections
Incubating with primary antibody for 1 hour at room temperature
Using secondary antibodies (e.g., anti-goat, 1:500) for 20 minutes
Scoring based on both staining intensity and proportion of positively stained cells
ELISA: For quantitative assessment of ITIH3 in serum samples, as demonstrated in studies examining ITIH3 as a biomarker in myasthenia gravis .
Mass Spectrometry: Proteomic approaches like iTRAQ (isobaric tag for relative and absolute quantitation) have been used to identify differences in ITIH3 expression levels in experimental models .
Successful immunoprecipitation (IP) of ITIH3 requires careful optimization of experimental conditions. Based on research protocols:
Antibody Selection: Use high-specificity anti-ITIH3 antibodies validated for IP applications. Researchers investigating ITIH3 in myasthenia gravis successfully employed immunoprecipitation of ITIH3 followed by proteomics to identify interaction partners .
Buffer Composition:
Lysis buffer: RIPA buffer containing protease inhibitors
Washing buffer: PBS with 0.1% Tween-20
Elution buffer: Glycine-HCl (pH 2.5-3.0) followed by immediate neutralization
Protocol Optimization:
Pre-clear lysates with protein A/G beads to reduce non-specific binding
Incubate antibodies with protein lysates overnight at 4°C with gentle rotation
Use appropriate negative controls (isotype-matched IgG)
Validate pull-down efficiency by western blotting using a different anti-ITIH3 antibody
Downstream Analysis: For identification of ITIH3 interaction partners, mass spectrometry-based proteomics has proven effective, as demonstrated in studies identifying proteins crucial for neuromuscular transmission that interact with ITIH3 .
Designing effective ITIH3 knockdown experiments requires careful consideration of several factors:
RNA Interference (RNAi) Design:
Target sequence selection: Design multiple siRNA/shRNA sequences targeting different regions of ITIH3 mRNA
Construct appropriate vectors for stable expression (e.g., lentiviral vectors with fluorescent markers)
Verify knockdown efficiency: Aim for >70% reduction in protein expression
Validation of Knockdown Efficiency:
Western blotting is the standard method for confirming ITIH3 protein reduction
In published studies, successful ITIH3 RNAi cell lines showed protein expression levels at approximately 30% of those in parent cell lines and negative controls
Fluorescence verification: Successful transduction can be confirmed by cell fluorescence rates >80%
Functional Assays Post-Knockdown:
In Vivo Model Development:
Proper controls are critical for reliable ITIH3 immunohistochemistry results:
Primary Controls:
Positive tissue controls: Include tissues known to express ITIH3 (e.g., normal liver)
Negative tissue controls: Include tissues known not to express ITIH3
No primary antibody control: Perform parallel staining omitting the primary antibody
Isotype control: Use non-specific antibody of the same isotype as the ITIH3 antibody
Technical Controls:
Independent evaluation by multiple pathologists (preferably blinded to the research)
Standard tissue processing and antigen retrieval methods
Consistent antibody concentrations and incubation times
Scoring System Standardization:
Validation of Results:
Confirm IHC findings with alternative methods (western blotting, qPCR)
Use multiple antibodies targeting different epitopes of ITIH3 when possible
ITIH3 has emerged as a significant factor in chemotherapy resistance, particularly affecting cisplatin (DDP) sensitivity in ovarian cancer:
Investigating the interaction between ITIH3 and Bcl-2 family proteins requires comprehensive methodological approaches:
Protein Expression Analysis:
Western blotting to quantify expression levels of:
Immunoprecipitation followed by western blotting to detect direct protein-protein interactions
Functional Assays:
Apoptosis analysis using Annexin V/PE staining and flow cytometry to assess the effect of ITIH3 expression on cell death following cisplatin treatment
Cell viability assays (CCK-8) to determine IC50 values in cells with different ITIH3 expression levels
Real-time cell analysis using systems like xCELLigence RTCA to monitor dynamic changes in cell proliferation following treatment
Pharmacological Intervention:
Use of specific inhibitors like the Bcl-2 inhibitor ABT-737 to determine if blocking anti-apoptotic proteins can reverse the effects of ITIH3 silencing
Dose-response experiments to establish optimal inhibitor concentrations
In Vivo Validation:
Evaluating ITIH3 as a predictive biomarker requires a systematic approach:
Optimizing ITIH3 detection as a biomarker in myasthenia gravis requires specialized approaches:
Serum Analysis Protocols:
Collection standardization: Use serum separator tubes and process within 2 hours
Storage conditions: Aliquot and store at -80°C to avoid repeated freeze-thaw cycles
ELISA validation: Optimize antibody concentrations and establish standard curves
Machine learning approaches have successfully identified ITIH3 as a potential serum biomarker reflective of disease activity
Correlation with Clinical Metrics:
Structural Validation:
Protein Interaction Studies:
Investigating ITIH3 at neuromuscular junctions presents unique methodological challenges:
Tissue Sampling and Processing:
Obtain muscle biopsies from appropriate anatomical sites
Utilize both frozen sections (for optimal antigen preservation) and paraffin-embedded sections
Employ specialized fixation protocols to preserve neuromuscular junction integrity
Co-localization Studies:
Use dual or triple immunofluorescence labeling with:
Anti-ITIH3 antibodies
Markers of post-synaptic apparatus (e.g., α-bungarotoxin for acetylcholine receptors)
Pre-synaptic markers (e.g., synaptophysin)
Analyze using confocal microscopy for precise localization
Functional Correlation:
Correlate ITIH3 localization patterns with electrophysiological parameters
Compare staining patterns between:
Patients with active disease
Patients in remission
Healthy controls
Assess changes in ITIH3 distribution following therapeutic interventions
Molecular Interaction Analysis:
Investigate ITIH3 interactions with specific components of neuromuscular transmission
Apply proximity ligation assays to validate protein-protein interactions in situ
Conduct laser capture microdissection followed by proteomics to analyze the neuromuscular junction-specific interactome
Discrepancies between ITIH3 mRNA and protein expression represent a common challenge requiring systematic troubleshooting:
Validation of Detection Methods:
Confirm specificity of primers for mRNA detection (qRT-PCR)
Validate antibody specificity using positive and negative controls
Employ multiple antibodies targeting different epitopes of ITIH3
Consider using different housekeeping genes/proteins as references
Post-transcriptional Regulation Assessment:
Investigate potential microRNA-mediated regulation of ITIH3
Assess mRNA stability using actinomycin D chase experiments
Examine alternative splicing patterns that might affect antibody recognition sites
Protein Stability Analysis:
Measure ITIH3 protein half-life using cycloheximide chase assays
Investigate proteasomal and lysosomal degradation pathways
Examine post-translational modifications that might affect antibody recognition
Contextual Interpretation:
Consider tissue/cell-specific regulatory mechanisms
Examine potential disease-specific alterations in ITIH3 processing
Account for temporal dynamics in gene expression versus protein accumulation
Ensuring antibody specificity is crucial for reliable ITIH3 detection across multi-omics approaches:
Antibody Validation Process:
Genetic approaches: Test antibodies in ITIH3 knockout/knockdown models
Peptide competition assays: Pre-incubate antibodies with specific peptides
Orthogonal validation: Compare results across multiple antibodies
Cross-reactivity assessment: Test against related ITIH family members
Application-Specific Optimization:
Western blotting: Optimize blocking agents, antibody dilutions, and incubation times
Immunohistochemistry: Compare different antigen retrieval methods
Immunoprecipitation: Test various lysis and washing buffers
Flow cytometry: Optimize fixation and permeabilization protocols
Multi-omics Integration Considerations:
Ensure consistent sample processing across platforms
Apply statistical methods to account for platform-specific biases
Validate key findings using orthogonal techniques
Implement appropriate normalization strategies for cross-platform comparisons
Reporting Standards:
Document detailed antibody information (manufacturer, catalog number, lot, dilution)
Report all validation experiments performed
Share negative control data alongside positive results
Acknowledge potential limitations in antibody specificity
Exploring ITIH3 as a therapeutic target requires a comprehensive research strategy:
Target Validation Approaches:
Conduct gain-of-function experiments: Overexpress ITIH3 in resistant cancer cell lines to determine if sensitivity to cisplatin can be restored
Perform structure-function analyses: Identify critical domains of ITIH3 involved in mediating chemosensitivity
Develop conditional knockout models to evaluate tissue-specific effects of ITIH3 modulation
High-Throughput Screening Strategies:
Design cell-based assays to identify compounds that modulate ITIH3 expression or activity
Develop reporter systems to monitor ITIH3 promoter activity
Utilize CRISPR activation/inhibition screens to identify regulators of ITIH3
Combination Therapy Approaches:
Translational Models:
Develop patient-derived xenograft models with varying ITIH3 expression levels
Establish organoid cultures to test interventions in more physiologically relevant systems
Design early-phase clinical trials with ITIH3 expression as a stratification factor
Advancing ITIH3 research in complex tissue microenvironments requires cutting-edge methodological approaches:
Spatial Transcriptomics and Proteomics:
Apply technologies like Visium, Slide-seq, or CODEX to visualize ITIH3 expression patterns in relation to other cell types
Develop multiplexed imaging approaches to simultaneously visualize ITIH3 and its interaction partners
Integrate single-cell sequencing with spatial information to map ITIH3 expression at cellular resolution
3D Culture Systems:
Utilize organoid models to study ITIH3 in more physiologically relevant contexts
Develop co-culture systems incorporating multiple cell types (e.g., tumor cells with stromal components)
Apply microfluidic organ-on-chip approaches to model dynamic interactions
In Situ Protein Interaction Mapping:
Implement proximity labeling techniques (BioID, APEX) to identify ITIH3 interaction partners in live cells
Apply FRET/BRET approaches to monitor real-time protein interactions
Develop conditional interaction screening methods to identify context-specific binding partners
Live Imaging Innovations:
Generate fluorescently tagged ITIH3 constructs for real-time visualization
Apply optogenetic approaches to manipulate ITIH3 function with spatial and temporal precision
Develop biosensors to monitor ITIH3-dependent signaling events in living systems
Current best practices for ITIH3 detection vary by application but should adhere to these general principles:
Antibody Selection and Validation:
Use antibodies validated for the specific application (western blotting, IHC, IP)
Verify specificity through appropriate controls (ITIH3 knockdown/knockout)
Consider using multiple antibodies targeting different epitopes
For IHC, the goat-anti-ITI-H3 (L-15) antibody (1:200; cat. no. sc-33949; Santa Cruz Biotechnology, Inc.) has been successfully employed in published research
Protocol Optimization by Application:
Western blotting: Standard protocols with GAPDH or Actin as loading controls
Immunohistochemistry: 4% paraformaldehyde fixation, paraffin embedding, and standardized scoring systems
ELISA: Validated for serum biomarker studies, particularly in myasthenia gravis research
Proteomics: iTRAQ approaches have successfully identified ITIH3 expression changes in drug resistance models
Data Interpretation Guidelines:
Consider tissue/disease-specific expression patterns
Interpret results in the context of relevant biological pathways (e.g., Bcl-2 family proteins for cancer studies)
Correlate with clinical parameters when available
Recognize the limitations of individual detection methods
Reporting Standards:
Document detailed methodological information to ensure reproducibility
Include all relevant controls in publications
Share raw data when possible to facilitate meta-analyses
Acknowledge potential limitations in antibody specificity or assay sensitivity
Integrating ITIH3 research into broader disease contexts requires strategic approaches:
Pathway Integration Strategies:
Map ITIH3 interactions within established signaling networks
Identify convergent pathways between ITIH3 and known disease mechanisms
For cancer research, focus on integrating ITIH3 within apoptotic regulation networks, particularly the Bcl-2 family pathway
For neuromuscular diseases, examine connections between ITIH3 and established neuromuscular junction components
Biomarker Development Pipeline:
Progress from discovery (proteomics) to validation (targeted assays)
Establish standardized detection methods for clinical laboratories
Determine appropriate reference ranges in healthy populations
Conduct prospective studies to validate predictive value
Therapeutic Target Assessment:
Translational Research Framework:
Establish clinically annotated biobanks with ITIH3 characterization
Develop preclinical models that recapitulate human ITIH3 biology
Create interdisciplinary collaborations spanning basic science to clinical application
Implement machine learning approaches to integrate ITIH3 data with other disease parameters