HTRA1 is a protease involved in cleaving extracellular matrix proteins, IGF-binding proteins, and proteoglycans, with roles in processes like TGF-β signaling regulation and retinal angiogenesis . The HRP-conjugated antibody facilitates the detection of HTRA1 via enzyme-linked immunosorbent assays (ELISA), Western blotting (WB), or immunohistochemistry (IHC). HRP catalyzes chromogenic or chemiluminescent reactions, enabling quantifiable or visualized antigen detection .
In preclinical studies, HTRA1 antibodies were developed as inhibitors for age-related macular degeneration (AMD). The anti-HtrA1 Fab inhibitor (Fab15H6.v4.D221) demonstrated potent proteolytic activity inhibition in rabbit and cynomolgus monkey models, with pharmacodynamic biomarkers like Dickkopf-related protein 3 validated in phase 1 clinical trials . While not HRP-conjugated, this research underscores the importance of HTRA1 antibodies in disease modeling.
HTRA1 levels in CSF correlate with multiple sclerosis (MS) progression and disability. A study employing ELISA-based detection (likely involving HRP-conjugated antibodies) reported a receiver operating characteristic (ROC) area under the curve (AUC) of 0.903 for distinguishing untreated relapsing-remitting MS (RRMS) from healthy controls . This highlights the antibody’s utility in neurodegenerative diagnostics.
Immunohistochemistry with anti-HTRA1 antibodies revealed colocalization with amyloid deposits in Alzheimer’s disease (AD) brains. HTRA1 expression was observed in astrocytes and cortical neurons, suggesting its role in Aβ degradation pathways . While the study used unconjugated antibodies, the findings inform HTRA1’s biological relevance.
Western Blotting: The ABIN7168919 antibody detects a 51 kDa band corresponding to HTRA1 in human placental lysates and HAP1 cell lines .
ELISA Sensitivity: Achieves 82.6% sensitivity and 100% specificity for detecting HTRA1 in CSF of RRMS patients .
Cross-Reactivity: Demonstrated specificity for HTRA1 without cross-reactivity with HtrA2 or HtrA3 in human brain samples .
HTRA1 is a multidomain serine protease that belongs to the high-temperature requirement A (HtrA) family. It contains an N-terminal insulin-like growth factor binding protein (IGFBP) domain, a Kazal-type trypsin inhibitor motif, a trypsin-like protease domain, and a C-terminal PDZ domain . HTRA1 is extensively studied due to its involvement in:
Age-related macular degeneration (AMD) pathogenesis and progression
Regulation of TGF-beta signaling pathways
Extracellular matrix protein degradation
Tumor suppression activities
Neurodegenerative diseases including Alzheimer's disease
Arthritis (rheumatoid and osteoarthritis)
Cerebral autosomal recessive arteriopathy (CARASIL)
HTRA1 functions as a protease that cleaves various substrates including extracellular matrix proteins like fibronectin, proteoglycans (aggrecan, decorin, fibromodulin), and insulin-like growth factor binding proteins . Through these activities, HTRA1 plays critical roles in tissue remodeling, growth factor availability, and cellular signaling.
When working with HTRA1 antibodies, understanding the protein's molecular characteristics is essential:
In Western blot applications, HTRA1 typically appears as a major band at approximately 51 kDa, with potential additional bands representing autolytic fragments or oligomeric forms .
HRP-conjugated HTRA1 antibodies offer several advantages for research applications:
Optimal working dilutions for HRP-conjugated HTRA1 antibodies typically range from 1:500-1:1000 for Western blot and 1:50-1:500 for immunohistochemistry applications, though researchers should perform optimization for their specific experimental conditions .
Proper controls are essential for validating experimental results with HTRA1 antibodies:
Positive Controls:
Negative Controls:
HTRA1 knockout HAP1 cell lysates (validated control for specificity)
Cell lines treated with HTRA1-specific siRNA (>90% reduction has been documented)
Primary antibody omission control
Isotype-matched irrelevant antibody control
For genetic validation, both HTRA1 knockout and knockdown approaches have been successfully employed, with knockout HAP1 cells showing complete loss of the expected 51 kDa band in Western blot applications .
Optimizing Western blot protocols for HTRA1 detection requires consideration of several technical factors:
Sample Preparation:
Test both reducing and non-reducing conditions (some epitopes may be masked under certain conditions)
Include protease inhibitors in lysis buffers to prevent HTRA1 autolysis
Load 20-30 μg of total protein for moderate HTRA1 expression
Protocol Optimization:
Block with 5% non-fat milk in TBST (optimal for reducing background)
Antibody dilution: Start with 1:500-1:1000 and adjust as needed
Use Immunoblot Buffer Group 1 for optimal results with certain antibodies
For challenging samples, consider overnight incubation at 4°C
Detection Strategies:
Test different exposure times to capture optimal signal without saturation
For low abundance detection, use enhanced chemiluminescence substrates
Consider testing antibodies that target different epitopes of HTRA1 (e.g., N-terminal vs. C-terminal)
A systematic approach to optimization will yield consistent and specific detection of HTRA1 protein in your experimental system.
Antibody validation is critical for ensuring reliable research outcomes. For HTRA1 antibodies, multiple validation approaches should be employed:
Genetic Validation:
Compare signal between wild-type and HTRA1 knockout cell lines (e.g., HAP1 knockout cell line)
Use HTRA1-specific siRNA to knockdown expression (validated approaches have achieved >90% reduction)
Overexpression studies with recombinant HTRA1
Analytical Validation:
Peptide competition assays using the immunizing peptide/protein
Test reactivity against specific domains/fragments of HTRA1
Mass spectrometry confirmation of immunoprecipitated proteins
Functional Validation:
Activity-based probe (ABP) competition assays to measure inhibition of HTRA1 activity
Substrate cleavage assays to confirm functionality
Abcam's HTRA1 antibody (ab274322) demonstrates robust validation through knockout cell testing, showing complete loss of the 51 kDa band in HTRA1 knockout HAP1 cells .
When encountering weak or absent signals with HTRA1 antibodies, implement this systematic troubleshooting workflow:
Verify Antibody Activity:
Sample Quality Assessment:
Ensure proper protein extraction (test different lysis buffers)
Check for protein degradation (add fresh protease inhibitors)
Test sample preparation methods (non-reducing vs. reducing conditions)
Protocol Optimization:
Increase antibody concentration (try 2-5× recommended dilution)
Extend primary antibody incubation time (overnight at 4°C)
Use more sensitive detection reagents
Increase protein loading (50-75 μg total protein)
Technical Considerations:
A methodical approach to troubleshooting will help identify the source of the problem and guide appropriate solutions.
HTRA1 can exhibit complex banding patterns that require careful interpretation:
Expected Patterns:
Primary band at approximately 51 kDa (full-length HTRA1)
Lower molecular weight bands (35-50 kDa) may represent autolytic cleavage products
Higher molecular weight bands (100-150 kDa) may indicate dimers or trimers
Interpretation Guidelines:
Autolysis: HTRA1 undergoes self-cleavage, generating multiple fragments. This is particularly evident in samples with high HTRA1 expression or extended storage .
Oligomerization: HTRA1 can form dimers and trimers, especially under non-reducing conditions, appearing as bands at 2-3× the monomeric molecular weight.
Post-translational Modifications: Glycosylation or other modifications may cause shifts in apparent molecular weight.
Sample Preparation Impact: Fresh vs. stored samples and reducing vs. non-reducing conditions significantly affect banding patterns.
For complex patterns, consider additional validation approaches such as mass spectrometry or antibodies targeting different epitopes to confirm band identity.
Studying HTRA1's role in AMD requires sophisticated experimental approaches:
Tissue Analysis:
Perform comparative immunohistochemistry on normal vs. AMD-affected retinal tissues
Quantify HTRA1 expression in different retinal layers
Co-localize HTRA1 with drusen deposits and disease markers
Substrate Identification:
Analyze HTRA1-specific substrates in ocular tissues using N-terminomics
Develop ELISAs for substrate cleavage products as biomarkers
Functional Assessment:
Combine with activity-based probes (ABPs) to study active HTRA1 in vitreous samples
Use ABP competition assays to measure inhibition by therapeutic antibodies
Track pharmacodynamic biomarkers like DKK3 cleavage products
Therapeutic Development:
Screen for HTRA1 inhibitory antibodies using activity assays
Test efficacy of antibodies like Fab15H6.v4.D221 on HTRA1 activity
Measure effects on substrate processing and disease progression
This multi-faceted approach provides comprehensive insights into HTRA1's contribution to AMD pathogenesis and potential therapeutic interventions.
Researchers can employ sophisticated methods to evaluate HTRA1 inhibition combining activity-based probes (ABPs) with antibody detection:
ABP Competition Assay Design:
Pre-incubate HTRA1 with test inhibitors/antibodies at various concentrations
Add HTRA1-specific ABP (e.g., with TAMRA fluorescent tag)
Analyze by SDS-PAGE with fluorescence detection
Calculate inhibition potency (IC50) relative to controls
Experimental Setup:
| Component | Concentration | Purpose |
|---|---|---|
| Recombinant HTRA1 | 50-100 nM | Target enzyme |
| Test antibody | Serial dilutions (1 nM - 5 μM) | Potential inhibitor |
| HTRA1-specific ABP | 200-500 nM | Activity reporter |
| Controls | - No HTRA1 - No inhibitor - Known inhibitor | Establish assay parameters |
Readout and Analysis:
Quantify fluorescent band intensity at 50 kDa (intact HTRA1)
Plot inhibition curves (% activity vs. inhibitor concentration)
Calculate IC50 values (35.9-51.0 nM reported for anti-HTRA1 Fab15H6.v4.D221)
This system has been successfully applied to measure HTRA1 inhibition in both buffer systems and complex biological matrices such as vitreous humor .
Comprehensive identification and validation of HTRA1 substrates requires integrating multiple techniques:
N-terminomics/TAILS Approach:
Incubate biological samples with active HTRA1 vs. inactive HTRA1 or with/without inhibitory antibodies
Identify neo-N-terminal peptides generated by HTRA1 cleavage using mass spectrometry
Compare peptide profiles to identify HTRA1-specific cleavage sites
Validate findings with synthetic peptide substrates
Western Blot Validation:
Incubate purified candidate substrates with HTRA1
Analyze cleavage patterns by Western blot using substrate-specific antibodies
Confirm HTRA1-specific cleavage by including inhibitory antibodies
Quantify cleavage efficiency using densitometry
In Vivo Substrate Validation:
Administer HTRA1 inhibitory antibodies to animal models (e.g., intravitreal injection)
Collect tissue samples (e.g., vitreous humor)
Analyze substrate cleavage using Western blot or ELISA
Compare substrate profiles in treated vs. control animals
Research has identified several validated HTRA1 substrates including:
Dickkopf-related protein 3 (DKK3)
Clusterin (CLU)
Retinol-binding protein 3 (RBP3)
Fibronectin
These substrates can serve as valuable biomarkers for HTRA1 activity in both preclinical and clinical studies.
Investigating HTRA1's impact on TGF-beta signaling requires careful experimental design:
Receptor-Level Analysis:
Treat cells with recombinant HTRA1 with/without inhibitory antibodies
Immunoprecipitate TGF-beta receptors using specific antibodies
Analyze receptor integrity/processing by Western blot with HRP-conjugated antibodies
Quantify receptor degradation as a function of HTRA1 activity
Signaling Pathway Monitoring:
Establish reporter cell lines (SMAD-responsive elements)
Modulate HTRA1 levels (overexpression, knockdown, antibody inhibition)
Stimulate with TGF-beta ligands
Measure pathway activation (phospho-SMAD levels, reporter activity)
Multiplex Analysis Design:
| Component | Detection Method | Readout |
|---|---|---|
| Total HTRA1 | Anti-HTRA1, HRP-conjugated | Protein expression levels |
| Active HTRA1 | Activity-based probe + Western blot | Enzymatic activity |
| TGF-β Receptors | Receptor-specific antibodies | Integrity/processing |
| SMAD2/3 | Phospho-specific antibodies | Pathway activation |
| Target Genes | qRT-PCR | Transcriptional response |
This integrated approach provides mechanistic insights into how HTRA1 regulates TGF-beta signaling at multiple levels, from receptor processing to downstream transcriptional responses.
Monitoring HTRA1 activity in vivo requires sophisticated methods that balance sensitivity with specificity:
Intravitreal Administration Study Design:
Administer anti-HTRA1 antibodies at various doses (0.001-6 mg per eye)
Collect vitreous humor at different time points (1-14 days)
Analyze HTRA1 activity ex vivo using activity-based probes
Track substrate cleavage products as biomarkers
Activity Assessment Methods:
Activity-Based Probe Analysis: Label collected samples with HTRA1-specific ABPs to quantify active enzyme
Substrate Cleavage Monitoring: Analyze DKK3 processing as a pharmacodynamic biomarker
Western Blot Analysis: Detect HTRA1 and substrate processing using HRP-conjugated antibodies
Species-Specific Considerations:
Rabbit models show sustained HTRA1 inhibition at 0.02-2 mg doses for at least 14 days
Cynomolgus monkeys exhibit complete HTRA1 inhibition at 0.02-6 mg doses throughout study duration
Lower doses (0.001-0.002 mg) show transient inhibition with recovery by day 14
These approaches have successfully demonstrated dose-dependent and time-dependent inhibition of HTRA1 activity in vivo, providing crucial pharmacodynamic data for therapeutic development.