HTRA1 Antibody refers to monoclonal or polyclonal antibodies designed to target and modulate the activity of High-Temperature Requirement A1 (HTRA1), a secreted serine protease encoded by the HTRA1 gene. HTRA1 regulates extracellular matrix (ECM) remodeling, angiogenesis, and transforming growth factor-β (TGF-β) signaling pathways . Dysregulation of HTRA1 is implicated in cerebral small vessel disease (CSVD), age-related macular degeneration (AMD), cancer, and autoimmune disorders . Therapeutic HTRA1 antibodies aim to inhibit its proteolytic activity or neutralize its pathological effects.
Dickkopf-related protein 3 (DKK3): Aqueous humor levels of cleaved DKK3 serve as a pharmacodynamic biomarker for HTRA1 inhibition .
HTRA1 mutations reduce protease activity, leading to uncontrolled TGF-β signaling and vascular fibrosis .
Anti-HtrA1 antibodies restore TGF-β regulation, mitigating ECM accumulation in cerebral arterioles .
Cancer: HTRA1 overexpression in tumors (e.g., breast, ovarian) correlates with metastasis; antibodies block ECM degradation and angiogenesis .
Primary Membranous Nephropathy (MN): HTRA1 is a novel podocyte antigen targeted by IgG4 autoantibodies .
Fab15H6.v4.D221: Humanized Fab fragment with picomolar affinity for HTRA1. Inhibits vitreal HTRA1 activity in rabbits and cynomolgus monkeys for ≥14 days post-intravitreal injection .
| Antibody Clone | Host | Target Domain | Applications |
|---|---|---|---|
| 15H6.v4.D221 | Humanized | Protease domain | AMD therapy |
| MAB2916 | Mouse | Full-length HTRA1 | Western blot, IHC |
| 55011-1-AP | Rabbit | Protease domain | WB, IF, IP |
Western Blot: Detects HTRA1 at ~51 kDa (non-reducing conditions) .
Immunohistochemistry (IHC): Localizes HTRA1 in vascular endothelia and podocytes .
Clinical Limitations: Phase II trials showed no efficacy in GA, highlighting incomplete understanding of HTRA1’s role in AMD .
Biomarker Optimization: DKK3 validation requires larger cohorts to confirm sensitivity .
Therapeutic Expansion: Targeting HTRA1 in TGF-β-driven fibrosis and autoimmune MN is under exploration .
HTRA1, also known as HTRA, PRSS11, and L56, belongs to the peptidase S1B family. It functions as a protease that regulates the availability of various proteins in cellular pathways. HTRA1 is expressed in multiple tissue types with significant expression observed in:
Human tissues: Placenta shows strong expression as demonstrated in immunohistochemistry studies
Cell lines: L02 cells, HAP1 cells, and HepG2 cells consistently show HTRA1 expression in Western blot analyses
Animal tissues: Mouse brain tissue exhibits detectable levels of HTRA1 protein
Vascular system: Vascular smooth muscle cells (VSMCs) express HTRA1, where it plays a crucial role in vascular maturation and homeostasis
Renal system: HTRA1 serves as a podocyte antigen in the glomerular system
HTRA1 has a calculated molecular weight of 51 kDa, though observed molecular weights in experimental settings may vary (35 kDa, 42 kDa, and 50 kDa) depending on post-translational modifications and experimental conditions .
HTRA1 antibodies have been validated for multiple research applications with specific dilution recommendations:
| Application | Recommended Dilution | Validated Sample Types |
|---|---|---|
| Western Blot (WB) | 1:500-1:2000 | L02 cells, mouse brain tissue, HAP1 cells, HepG2 cells |
| Immunoprecipitation (IP) | 0.5-4.0 μg for 1.0-3.0 mg of total protein lysate | L02 cells, HAP1 cells |
| Immunohistochemistry (IHC) | 1:50-1:500 | Human placenta tissue |
| Immunofluorescence (IF/ICC) | 1:10-1:100 | HepG2 cells |
| ELISA | Application-dependent | Various sample types |
It is recommended to titrate the antibody in each testing system to obtain optimal results, as the effective concentration may be sample-dependent .
For optimal preservation of antibody activity:
Store at -20°C in aliquots to avoid repeated freeze-thaw cycles
HTRA1 antibodies in PBS with 0.02% sodium azide and 50% glycerol (pH 7.3) are typically stable for one year after shipment when properly stored
Aliquoting is generally unnecessary for -20°C storage
Small volume formats (20μl) may contain 0.1% BSA as a stabilizing agent
To ensure experimental validity:
Positive controls: Use L02 cells, HAP1 cells, HepG2 cells, or mouse brain tissue for Western blot applications
Negative controls: HTRA1 knockout or knockdown samples provide optimal negative controls, as demonstrated in studies using HtrA1-knockout mice for antibody development
Specificity verification: Compare HTRA1 wild-type samples with HtrA1 serine-to-alanine catalytically inactive mutants to confirm specificity in activity-based assays
Cross-reactivity assessment: Test against human, mouse, and rat samples, as the antibody shows documented reactivity with these species
HTRA1 antibodies have shown significant potential in therapeutic development, particularly for age-related macular degeneration (AMD):
Development approach: Anti-HtrA1 antibodies can be obtained by immunizing HtrA1-knockout mice with recombinant HtrA1 protease domain (HtrA1-PD)
Optimization process: The therapeutic development pipeline typically includes:
Initial hybridoma screening (identifying clone 15H6 with strong inhibition of HtrA1 enzymatic activity)
Humanization through CDR grafting into human consensus frameworks
Modification of problematic residues (N94A for cleavage, D55 for isomerization)
Affinity maturation via Fab phage display with deep-sequencing analysis
Clinical application: The development of HtrA1-blocking Fab fragments has enabled testing of the therapeutic hypothesis that HtrA1 protease activity contributes to AMD progression
Significance: Genome-wide association studies have identified genetic variation at the ARMS2/HTRA1 locus as a risk factor for AMD development and progression, making HtrA1 a relevant therapeutic target
Confirming target engagement is crucial for validating anti-HTRA1 antibody efficacy:
Activity-based probes (ABPs):
Development of HtrA1-directed ABPs using diphenyl phosphonate as the reactive group targeting the nucleophilic active-site serine residue
Incorporation of Val and Leu at P1 and P2 positions to direct ABP reactivity against HtrA1
Addition of carboxytetramethylrhodamine (TAMRA) through a polyethylene glycol linker as a fluorescent reporter
N-terminomic proteomic profiling:
Pharmacodynamic biomarkers:
Dickkopf-related protein 3 (DKK3) has been identified as a robust biomarker for anti-HtrA1 activity
This biomarker can be measured in both preclinical animal models and clinical samples
Successfully employed in a phase 1 study of geographic atrophy patients to demonstrate anti-HtrA1 Fab activity and duration
HTRA1 functions as a critical regulator of multiple signaling pathways:
TGFβ signaling regulation:
Notch signaling modulation:
The Notch ligand JAG1 is a substrate for HTRA1
HTRA1 cleaves JAG1 in the cytosol, leading to rapid degradation of the remaining JAG1 protein
JAG1/NOTCH3 signaling is crucial for differentiation, maintenance, and contractility of vascular smooth muscle cells
Synergistic effects:
TGFβ and Notch pathways synergistically stimulate expression of HES and HEY transcriptional repressors
Moderate overexpression of NOTCH3-ICD increases SM22α protein levels
Higher NOTCH3-ICD expression decreases SM22α expression, indicating hyper-activation of a negative feedback loop
Combined overexpression of HES1 and HEYL represses transcription of α-SMA and SM22α in human umbilical artery smooth muscle cells
HTRA1 antibodies serve as valuable tools in investigating various disease states:
Age-related macular degeneration (AMD):
Membranous nephropathy (MN):
HTRA1 has been identified as a novel podocyte antigen in a subset of patients with primary MN
Patient sera react by immunoblotting with a 51-kD protein within glomerular extracts
Anti-HTRA1 antibody titers appear to correlate with disease course
Serial monitoring of anti-HTRA1 antibodies could facilitate diagnostic and therapeutic decisions in this demographic group (mean age 67.3 years)
Vascular disorders:
Loss of HTRA1 in vascular smooth muscle cells (VSMCs) impairs their maturation and function
HTRA1 is essential for vascular homeostasis through its regulation of Notch and TGFβ signaling
HTRA1 antibodies can help investigate the role of this protease in familial small vessel disease, where blood vessel functions are impaired
When investigating HTRA1 substrates, researchers should consider:
N-terminomics approach:
Activity-based profiling:
Cross-species validation:
Biomarker identification:
For effective immunohistochemical detection of HTRA1:
Primary recommendation: Antigen retrieval with TE buffer at pH 9.0
Alternative method: Antigen retrieval with citrate buffer at pH 6.0
Validated tissue: Human placenta tissue shows positive IHC results
Recommended dilution: 1:50-1:500, with optimization for specific tissue types
When analyzing HTRA1 expression:
Expected molecular weights:
Verification strategy:
For cross-species HTRA1 research:
Validated reactivity: HTRA1 antibodies have been tested and confirmed to react with human, mouse, and rat samples
Cited reactivity: Published literature reports additional reactivity with zebrafish samples
Cross-species validation: Important substrates of HTRA1 have been identified through cross-species studies, suggesting conservation of function
Knockout models: HtrA1-knockout mice have been generated by traditional homologous recombination methods and serve as valuable tools for specificity controls and comparative studies
To properly assess HTRA1 inhibition by antibodies:
Enzymatic activity controls:
Substrate processing:
Signaling pathway analysis:
To improve specificity:
Antibody selection: Use antibodies derived from HtrA1-knockout animals for immunization, which reduces background reactivity
Purification method: Antigen affinity-purified antibodies offer higher specificity
Validation approach: Confirm antibody specificity using multiple techniques (WB, IP, IHC, IF/ICC) and positive/negative controls
Buffer optimization: PBS with 0.02% sodium azide and 50% glycerol at pH 7.3 provides optimal stability and specificity
Performance variations may stem from:
Sample preparation: Different lysis buffers, fixation methods, or antigen retrieval techniques can affect epitope accessibility
Protein conformation: Native versus denatured conditions influence epitope exposure
Background reduction: Increasing blocking agent concentration or adjusting antibody dilution can improve signal-to-noise ratio
Application-specific considerations:
WB: Optimize transfer conditions and blocking agents
IHC: Test alternative antigen retrieval methods (TE buffer pH 9.0 versus citrate buffer pH 6.0)
IP: Adjust antibody-to-lysate ratio (0.5-4.0 μg antibody for 1.0-3.0 mg total protein)
IF/ICC: Consider permeabilization methods and fixation protocols
When encountering variability:
Antibody titration: Test multiple dilutions to determine optimal concentration for each application and sample type
Controls expansion: Include both positive controls (L02 cells, HAP1 cells, HepG2 cells, mouse brain tissue) and negative controls (HTRA1 knockdown/knockout samples)
Cross-validation: Confirm results using alternative detection methods or antibody clones
Activity verification: For functional studies, use activity-based probes to confirm that observed effects correlate with HTRA1 enzymatic activity
HTRA1 antibodies have facilitated significant discoveries:
AMD pathogenesis: Anti-HtrA1 Fab inhibitors have demonstrated that HtrA1 proteolytic activity contributes to AMD progression
Membranous nephropathy: Identification of HTRA1 as a novel podocyte antigen has expanded our understanding of this glomerular disease
Vascular disorders: HTRA1 inhibition studies have revealed its role in vascular maturation and homeostasis through Notch and TGFβ signaling regulation
The translational potential includes:
Diagnostic biomarkers: Anti-HTRA1 antibody titers may serve as biomarkers in membranous nephropathy, facilitating diagnosis and treatment decisions
Therapeutic development: HtrA1-blocking antibodies show promise for treating AMD, with pharmacodynamic biomarkers like DKK3 enabling monitoring of therapeutic efficacy
Personalized medicine: Genetic variation at the ARMS2/HTRA1 locus may inform patient selection for anti-HTRA1 therapies in AMD