HTRA2 antibodies are employed in diverse experimental workflows to study mitochondrial dynamics, apoptosis, and disease mechanisms.
Detection of HTRA2 in Cell Lysates: Used to assess protein expression levels in cancer cells (e.g., PC-12, Jurkat, HeLa) and neuronal tissues .
Stress-Induced Processing: Monitors cleavage during mitochondrial stress, as seen in malaria parasites and neurodegenerative models .
Localization Studies: Identifies HTRA2 translocation from mitochondria to cytosol during apoptosis (e.g., staurosporin-treated Jurkat cells) .
Neuroprotection Models: Investigates HTRA2’s role in glaucoma and Alzheimer’s disease using retinal explants .
Protease Activity Inhibition: Antibodies like CDR1 peptide (ASGYTFTNYGLSWVR) inhibit HTRA2’s proteolytic function, tested via co-immunoprecipitation (Co-IP) and mass spectrometry .
Autophagy Regulation: Links HTRA2 to autophagic pathways by analyzing interactions with Beclin-1 and HAX-1 .
HTRA2 antibodies have elucidated the protein’s roles in apoptosis, senescence, and immune regulation.
Caspase Activation: HTRA2 cleaves inhibitor of apoptosis proteins (IAPs), promoting caspase-dependent apoptosis .
Neurodegeneration: Loss of HTRA2 protease activity causes mitochondrial dysfunction and neuronal death, as shown in mnd2 mice .
Radiation-Induced Senescence: HTRA2 mediates sustained proliferation arrest and SA-β-Gal staining in lung cancer cells .
Vimentin Cleavage: HTRA2 disassembles vimentin filaments, contributing to senescence-associated phenotypes .
Rheumatoid Arthritis (RA): HTRA2 suppresses Th17 differentiation and IL-17 production by degrading STAT3, reducing CIA severity in mice .
Therapeutic Potential: Overexpression of HTRA2 attenuates joint inflammation and cartilage damage in RA models .
Glaucoma Models: CDR1 peptide binds HTRA2, inhibiting its protease activity and protecting retinal ganglion cells .
Alzheimer’s Disease: HTRA2 promotes autophagy by degrading HAX-1, enhancing clearance of amyloid plaques .
PAT4G8AT.
HTRA2 (also known as OMI or PRSS25) is a mitochondrial serine protease with versatile biological functions ranging from apoptosis regulation to maintaining neuronal cell survival and mitochondrial homeostasis. The protein contains an N-terminal mitochondrial targeting sequence (MTS), a transmembrane domain (TM), a central protease domain, a C-terminal PDZ domain, and an unconventional IAP-binding motif . HTRA2 is particularly significant in research because:
Loss of HTRA2 protease function is associated with neurodegeneration, while overactivation of its proteolytic function is linked to cell death and inflammation . Studies have shown increased HTRA2 protein activity in brain tissues of Alzheimer's disease patients, suggesting a neuroprotective role through enhancement of autophagic processes . Furthermore, HTRA2 promotes the degradation of mutant proteins like A53T α-synuclein through autophagy and might be involved in amyloid plaque removal . The mnd2 (motor neuron degeneration) mice with inactivating mutations in the HTRA2 protease domain exhibit muscle wasting and neurodegeneration, while HtrA2 knockout mice display neuronal degeneration and a parkinsonian phenotype .
These diverse functions and disease associations make HTRA2 a compelling target for researchers studying neurodegenerative diseases, apoptosis, and mitochondrial function.
Various HTRA2 antibodies are available targeting different regions of the protein and optimized for different experimental applications. Selection should be based on the specific research needs:
Antibody Types by Target Region:
C-Terminal targeting antibodies (e.g., antibodies against AA 278-458, AA 334-458, AA 359-458)
N-Terminal targeting antibodies (e.g., antibodies against AA 73-102)
Mid-region targeting antibodies (e.g., antibodies against AA 231-330)
Antibody Types by Host and Format:
When selecting an antibody, researchers should consider:
The specific epitope being targeted (which may affect detection of different HTRA2 isoforms)
The experimental application (WB, IHC, IP, ELISA, IF)
Species reactivity (human, mouse, rat)
The format (conjugated vs. unconjugated)
For example, when studying the cleaved, active form of HTRA2 released from mitochondria, C-terminal antibodies may be more appropriate as this region remains intact after processing. For co-immunoprecipitation studies, antibodies validated for IP applications should be selected .
Validating antibody specificity is crucial for obtaining reliable results. For HTRA2 antibodies, consider these methodological approaches:
Positive and Negative Controls:
Positive control: Use tissues or cell lines known to express HTRA2 (most human cells express HTRA2, with higher levels in metabolically active tissues)
Negative control: Use HTRA2 knockout cells/tissues where available, or tissues known to have very low expression
Molecular Weight Verification:
Full-length HTRA2 is approximately 46 kDa before mitochondrial import
Upon mitochondrial import and processing, HTRA2 is cleaved to yield products of approximately 37 and 35 kDa
Verify that your antibody detects bands at these expected molecular weights
Peptide Competition Assay:
Pre-incubate the antibody with the immunizing peptide (if available)
Loss of signal in Western blot or immunostaining confirms specificity
siRNA/shRNA Knockdown:
Transfect cells with HTRA2-specific siRNA/shRNA
Reduction in signal intensity proportional to knockdown efficiency confirms specificity
Multiple Antibody Verification:
Use multiple antibodies targeting different epitopes of HTRA2
Consistent results across different antibodies increase confidence in specificity
Each validation method provides complementary information, and combining multiple approaches provides the strongest evidence for antibody specificity.
HTRA2 has been implicated in several neurodegenerative diseases, particularly Parkinson's disease (PD) and Alzheimer's disease (AD). HTRA2 antibodies can be instrumental in investigating these connections through various methodological approaches:
Tissue Expression Analysis:
Immunohistochemistry (IHC) using HTRA2 antibodies can reveal altered expression patterns in neurodegenerative disease tissues compared to healthy controls
Quantitative Western blotting can measure HTRA2 expression levels in different brain regions affected by neurodegenerative diseases
Post-translational Modification Analysis:
Phospho-specific antibodies can detect specific modifications of HTRA2 associated with disease states
Co-immunoprecipitation followed by mass spectrometry can identify disease-specific modifications
Protein-Protein Interaction Studies:
Co-immunoprecipitation using HTRA2 antibodies can identify interaction partners in disease contexts
Research has shown that HTRA2 has complex protein interaction networks in neurological tissues, with many interactors involved in ER to Golgi anterograde transport (e.g., AP3D1), aggrephagy (e.g., PSMC1), and pyruvate metabolism/citric acid cycle (e.g., SHMT2)
Changes in these interaction networks in disease states may reveal pathological mechanisms
Subcellular Localization Studies:
Immunofluorescence with HTRA2 antibodies can detect abnormal translocation from mitochondria in disease models
This is particularly relevant as HTRA2 release from mitochondria is associated with apoptotic signaling
In Vivo and Ex Vivo Models:
HTRA2 antibodies can be used to characterize disease phenotypes in animal models of neurodegeneration
For example, HtrA2 null mutants exhibit mild mitochondrial defects, loss of flight and climbing ability, and sensitivity to oxidative stress and mitochondrial toxins, sharing phenotypic characteristics with Drosophila models of PD
These approaches can provide insights into how HTRA2 dysfunction contributes to neurodegenerative processes and identify potential therapeutic targets.
Studying HTRA2 protease activity is crucial for understanding its biological functions. Several methodologies can be used, with antibodies playing complementary roles:
Fluorogenic Substrate Assays:
HTRA2 protease activity can be measured using fluorogenic peptide substrates such as the H2-optimal substrate
This substrate contains a quencher and fluorophore unit, where cleavage disrupts the quencher/fluorophore complex, allowing fluorescence measurement
The assay is typically performed with recombinant HTRA2 (100 nM) in a protease assay buffer (50 mM TRIS, 0.5 mM EDTA, 1 mM DTT, pH 8.0) containing 10 μM H2-optimal substrate
Fluorescence is monitored over time to determine the enzymatic reaction rate
Protease Inhibitor Studies:
Inhibitors like UCF-101 (typically used at 30 μM) specifically target the catalytic domain of HTRA2
Synthetic peptides like ASGYTFTNYGLSWVR (CDR1 peptide) have been shown to inhibit HTRA2 activity
Antibodies can be used to verify inhibitor specificity through co-immunoprecipitation experiments
Co-immunoprecipitation of HTRA2 Substrates:
HTRA2 antibodies can be used to immunoprecipitate HTRA2 along with its bound substrates
Mass spectrometry analysis of the immunoprecipitated complexes can identify novel substrates
Changes in substrate profiles under different conditions (e.g., disease states, stress conditions) can reveal context-specific functions
In-gel Zymography:
HTRA2 can be separated by non-denaturing PAGE containing a substrate (e.g., casein)
After electrophoresis, the gel is incubated to allow proteolysis, then stained to visualize areas of substrate degradation
Western blotting with HTRA2 antibodies on parallel gels can confirm the identity of the proteolytic bands
Cellular Assays:
Immunocytochemistry with HTRA2 antibodies can detect HTRA2 translocation from mitochondria during apoptosis
Live-cell imaging combined with immunofluorescence can track dynamic changes in HTRA2 localization and correlate with protease activity
These complementary approaches provide a comprehensive understanding of HTRA2 protease activity in different contexts.
Co-immunoprecipitation (Co-IP) is a powerful technique to study HTRA2 protein interactions. Based on published methodologies, here are optimized protocols and considerations:
Antibody Selection:
Choose antibodies specifically validated for immunoprecipitation (IP)
Consider using antibodies targeting different epitopes for confirmation, as some interactions might be masked by antibody binding
For tagged recombinant HTRA2, consider tag-specific antibodies (e.g., His-tag antibodies for 6xHis-tagged HTRA2)
Sample Preparation:
For tissue samples: Homogenize tissue in a gentle lysis buffer (e.g., PBS with protease inhibitors) to preserve protein-protein interactions
For cell cultures: Lyse cells in non-denaturing buffers (e.g., RIPA buffer with reduced detergent concentration)
Maintain low temperatures (4°C) throughout to preserve interactions
Include protease and phosphatase inhibitors to prevent degradation and modification of interaction partners
Pre-clearing:
Pre-clear lysates with protein A/G beads to reduce non-specific binding
This step is especially important when working with complex tissue samples like brain or retina
Immunoprecipitation:
For direct IP: Incubate clarified lysate with HTRA2 antibody (typically 2-5 μg per mg of total protein) followed by protein A/G beads
For recombinant His-tagged HTRA2: Spike recombinant protein (e.g., 2 μg) into homogenized tissue (e.g., 5 mg) and capture using Ni-NTA magnetic beads (e.g., 40 μL)
Incubate overnight at 4°C with gentle rotation to maximize binding while minimizing degradation
Washing and Elution:
Wash beads 2-4 times with PBS or appropriate buffer to remove non-specific binders
For His-tagged HTRA2, elute bound proteins by pH shift or with imidazole
For antibody-based IP, elute with low pH buffer or by boiling in SDS sample buffer
Controls:
Negative control: Perform parallel IP with isotype-matched control antibody or with lysate not containing HTRA2
Specificity control: Include conditions with HTRA2 inhibitors (e.g., UCF-101) or competitors (e.g., synthetic CDR peptide) to identify specific versus non-specific interactions
Analysis:
Analyze eluates by mass spectrometry for unbiased interaction partner identification
Confirm key interactions by Western blotting with specific antibodies
Compare interaction profiles under different conditions (e.g., with/without inhibitors, disease versus healthy state)
Following this optimized protocol can reveal complex protein interaction networks of HTRA2 in various tissues and conditions.
Immunohistochemistry (IHC) with HTRA2 antibodies in neurodegenerative disease tissues requires special considerations to obtain reliable and informative results:
Tissue Fixation and Processing:
HTRA2 is a mitochondrial protein that can relocalize during apoptosis, so fixation method is critical
For formalin-fixed paraffin-embedded (FFPE) tissues, optimize fixation time to prevent overfixation which may mask epitopes
For frozen sections, use gentle fixation (e.g., 4% paraformaldehyde for 10-15 minutes) to preserve antigenicity
Consider using both FFPE and frozen section approaches as complementary methods
Antigen Retrieval:
HTRA2 epitopes may be masked during fixation, requiring antigen retrieval
Test both heat-induced epitope retrieval (HIER) methods (e.g., citrate buffer pH 6.0, EDTA buffer pH 9.0) and enzymatic methods (e.g., proteinase K)
Optimize retrieval conditions for your specific antibody and tissue type
Antibody Selection and Validation:
Select antibodies specifically validated for IHC applications
Validate antibody specificity in your tissue of interest using positive and negative controls
Consider using multiple antibodies targeting different epitopes to confirm staining patterns
Background Reduction:
Block endogenous peroxidase activity (for HRP-based detection) or endogenous biotin (for biotin-based detection)
Use tissue-matched blocking serum containing the same species as the secondary antibody
Include detergents (e.g., 0.1-0.3% Triton X-100) in antibody diluents to reduce non-specific membrane binding
Consider using specialized blocking reagents for tissues with high background (e.g., brain tissues)
Signal Amplification and Detection:
For low abundance targets, use signal amplification methods (e.g., tyramide signal amplification)
When studying co-localization, use fluorescent secondary antibodies with spectrally distinct fluorophores
For quantitative analysis, use automated image analysis software with appropriate controls
Interpretation Challenges in Neurodegenerative Tissues:
Neurodegenerative tissues often contain protein aggregates that can trap antibodies non-specifically
Include controls using pre-immune serum or isotype control antibodies
Compare staining patterns with other mitochondrial markers to confirm subcellular localization
When studying diseased tissue, include age-matched controls and analyze multiple brain regions
Quantification Approaches:
Use digital image analysis for objective quantification of HTRA2 expression levels
Normalize HTRA2 staining to appropriate housekeeping proteins or mitochondrial markers
Consider semi-quantitative scoring systems (e.g., H-score) for comparative studies across disease stages
These technical considerations will help ensure reliable and informative results when using HTRA2 antibodies for IHC in neurodegenerative disease tissues.
HTRA2 belongs to the HtrA family of serine proteases, which in humans includes HTRA1, HTRA2, HTRA3, and HTRA4. Designing experiments to assess potential cross-reactivity requires systematic approaches:
Sequence Analysis and Epitope Mapping:
Perform sequence alignment of all HtrA family members to identify regions of high homology
Map the antibody epitope location and assess homology with corresponding regions in other HtrA proteins
Use epitope prediction algorithms to identify potential cross-reactive epitopes
Recombinant Protein Testing:
Express and purify all HtrA family members as recombinant proteins
Perform Western blot analysis with the HTRA2 antibody against all family members
Quantify relative binding affinity to determine cross-reactivity levels
Knockout/Knockdown Validation:
Use HTRA2 knockout or knockdown cell lines as negative controls
Test whether the antibody produces signals in these systems (which would indicate cross-reactivity)
Additionally, overexpress other HtrA family members in these systems to directly assess cross-reactivity
Immunoprecipitation-Mass Spectrometry:
Perform immunoprecipitation with the HTRA2 antibody
Analyze the precipitated proteins by mass spectrometry
Identify any other HtrA family members in the precipitate
Immunohistochemistry with Specific Controls:
Perform IHC on tissues with known expression patterns of different HtrA family members
Compare with IHC using validated antibodies specific for other HtrA proteins
Include tissues from HTRA2 knockout animals as controls
Peptide Competition Assays:
Design peptides corresponding to the epitope regions of all HtrA family members
Perform competition assays to determine whether peptides from other family members can block antibody binding
Reporting Standards:
Document all cross-reactivity testing methodologies in publications
Provide quantitative assessments of cross-reactivity where possible
Clearly state the limitations of the antibody regarding potential cross-reactivity
This systematic approach will provide comprehensive information about potential cross-reactivity, allowing researchers to make informed decisions about antibody applications and data interpretation.
HTRA2's localization in mitochondria and its role in mitochondrial homeostasis make it an important target for studying mitochondrial function. Here are optimized protocols using HTRA2 antibodies:
Subcellular Fractionation and Western Blotting:
Isolate mitochondrial, cytosolic, and nuclear fractions using differential centrifugation
Verify fraction purity using markers like VDAC (mitochondria), GAPDH (cytosol), and Lamin B (nucleus)
Perform Western blotting with HTRA2 antibodies to track HTRA2 localization under normal and stress conditions
Quantify the ratio of mitochondrial to cytosolic HTRA2 as an indicator of mitochondrial release
Mitochondrial Morphology Assessment:
Perform immunofluorescence co-staining with HTRA2 antibodies and mitochondrial markers (e.g., MitoTracker, TOM20)
Use confocal microscopy to analyze mitochondrial morphology parameters (length, fragmentation, network connectivity)
Compare these parameters in cells with normal versus altered HTRA2 expression/activity
HtrA2 knockout mice exhibit abnormal mitochondria, suggesting HTRA2's importance in maintaining mitochondrial integrity
Mitochondrial Membrane Potential:
Use potential-sensitive dyes (e.g., JC-1, TMRM) in combination with immunofluorescence for HTRA2
Correlate HTRA2 expression/localization with membrane potential changes
Perform time-lapse imaging to track dynamic changes during cellular stress
Mitochondrial Respiration Analysis:
Measure oxygen consumption rate (OCR) using platforms like Seahorse XF Analyzer
Compare OCR in cells with normal, depleted, or inhibited HTRA2
Use HTRA2 antibodies to verify HTRA2 status in parallel samples
HtrA2 mutants exhibit mitochondrial defects that could affect respiratory function
Mitochondrial Stress Response:
Induce mitochondrial stress with toxins like rotenone or CCCP
Track HTRA2 expression, processing, and localization using specific antibodies
Correlate with markers of mitochondrial unfolded protein response (UPRmt)
HtrA2 null mutants show sensitivity to oxidative stress and mitochondrial toxins
Mitophagy Assessment:
Co-stain for HTRA2 and mitophagy markers (e.g., PINK1, Parkin, LC3)
Track co-localization during mitophagy induction
Use live-cell imaging to monitor dynamics of HTRA2-positive mitochondria during mitophagy
Electron Microscopy with Immunogold Labeling:
Perform immunogold labeling with HTRA2 antibodies for transmission electron microscopy
Analyze ultrastructural localization of HTRA2 within mitochondria
Compare mitochondrial ultrastructure in normal versus disease models
These protocols provide complementary approaches to study HTRA2's role in mitochondrial function and dysfunction, particularly relevant for neurodegenerative disease research.
Western blotting with HTRA2 antibodies can present several challenges. Here are methodological solutions to common problems:
Potential causes and solutions:
Insufficient HTRA2 expression: Confirm HTRA2 expression in your sample using RT-PCR or enriched mitochondrial fractions
Antibody dilution too high: Perform titration experiments to determine optimal antibody concentration
Inefficient protein transfer: Verify transfer efficiency with Ponceau S staining; consider optimizing transfer conditions for mitochondrial proteins
Epitope masking: Try different extraction buffers; HTRA2 is a mitochondrial protein that may require specialized extraction methods
Epitope destruction during processing: Avoid excessive sample heating; include protease inhibitors in all buffers
Potential causes and solutions:
Cross-reactivity: Verify specificity using HTRA2 knockdown/knockout controls
HTRA2 processing products: HTRA2 exists in different forms (full-length ~46 kDa; processed forms ~37 and 35 kDa) ; confirm band identity using recombinant standards
Protein degradation: Include fresh protease inhibitors in all buffers; keep samples cold throughout preparation
Non-specific secondary antibody binding: Include proper blocking agents; test alternative secondary antibodies
Potential causes and solutions:
Insufficient blocking: Increase blocking time/concentration; test alternative blocking agents (e.g., BSA vs. milk)
Antibody concentration too high: Dilute primary and/or secondary antibodies
Membrane overexposure: Reduce exposure time during imaging
Detergent concentration: Optimize wash buffer detergent concentration (typically 0.05-0.1% Tween-20)
Potential causes and solutions:
HTRA2 expression variability: Standardize culture/treatment conditions; use appropriate housekeeping controls
Antibody batch variation: Use the same lot when possible; include positive controls with each experiment
Sample preparation inconsistency: Standardize protein extraction and quantification protocols
Loading control issues: Verify equal loading with total protein stains (e.g., Ponceau S, REVERT)
Potential causes and solutions:
Different epitopes: Different antibodies may recognize different forms of HTRA2; map the epitopes to understand what each antibody should detect
Isoform specificity: Verify which isoforms each antibody can detect
Post-translational modifications: Some antibodies may be sensitive to modifications that mask epitopes
Specificity differences: Validate each antibody using knockdown/knockout controls
Recommended optimization strategy:
Start with positive control samples known to express HTRA2 (e.g., HeLa cells)
Use mitochondrial enrichment to increase signal-to-noise ratio
Test multiple antibody dilutions (typically 1:500 to 1:5000)
Include appropriate molecular weight markers to identify specific HTRA2 forms
Document all conditions systematically to identify optimal parameters
Following these troubleshooting strategies will help researchers obtain reliable and reproducible Western blot results with HTRA2 antibodies.
Contradictory results between different HTRA2 antibodies in neurodegenerative disease studies require systematic investigation and careful interpretation:
Sources of Contradictions:
Epitope-specific differences:
Different antibodies target different regions of HTRA2
Some epitopes may be masked or modified in disease states
Solution: Map the exact epitopes of each antibody to understand what each is detecting
Disease-specific modifications:
Neurodegenerative diseases may induce post-translational modifications or conformational changes
These changes may affect epitope accessibility differentially
Solution: Use techniques like mass spectrometry to identify disease-specific modifications
Processing differences:
Subcellular localization changes:
HTRA2 can relocalize from mitochondria during stress/disease
Solution: Use subcellular fractionation and localization studies with multiple antibodies
Methodological Approach to Resolve Contradictions:
Comprehensive antibody validation:
Validate each antibody using knockout/knockdown controls
Confirm specificity with peptide competition assays
Test reactivity against recombinant HTRA2 protein
Multi-technique confirmation:
Use complementary techniques (Western blot, IHC, IP-MS)
Compare results across techniques for each antibody
Consider that some antibodies may work better in certain applications
Control experiments:
Careful sample handling:
Standardize tissue collection, processing, and storage
Process disease and control samples identically
Document post-mortem intervals for human samples
Interpretation Framework:
When antibodies targeting different domains show different results:
Consider domain-specific modifications or interactions
Investigate whether disease-associated mutations affect specific domains
Examine whether proteolytic processing differs in disease tissues
When different techniques yield contradictory results:
Consider technique-specific limitations (e.g., epitope masking in FFPE tissues)
Evaluate whether sample preparation affects HTRA2 structure differently
Determine which technique provides the most relevant biological context
When results differ between disease models and human samples:
Consider species-specific differences in HTRA2 sequence and processing
Evaluate whether model systems accurately recapitulate human disease aspects
Assess whether disease duration/severity affects results
Data reporting best practices:
Report results from multiple antibodies separately rather than combining
Clearly document which antibody was used for each experiment
Discuss limitations and potential reasons for contradictions
Consider publishing null or contradictory results to advance the field
By following this systematic approach, researchers can transform contradictory results into valuable insights about disease-specific changes in HTRA2 biology.
HTRA2, also known as Omi, is a serine protease located in the mitochondria. It plays a crucial role in the regulation of apoptosis, a process of programmed cell death essential for maintaining cellular homeostasis. The HTRA2 protein is encoded by the HTRA2 gene in humans and has homologs in other species, including mice and rats.
HTRA2 is a mitochondrial protein that is released into the cytoplasm in response to apoptotic stimuli. It contains a serine protease domain and a PDZ domain, which are essential for its proteolytic activity and protein-protein interactions, respectively. The mature form of HTRA2 has a conserved IAP-binding motif (AVPS) at its N-terminus, which allows it to interact with and inhibit Inhibitor of Apoptosis Proteins (IAPs) .
HTRA2 is involved in the mitochondrial apoptotic pathway. Upon induction of apoptosis, HTRA2 is released from the mitochondria into the cytoplasm, where it binds to IAPs and neutralizes their anti-apoptotic effects. This interaction promotes the activation of caspases, the executioners of apoptosis, leading to cell death .
Mouse anti-human HTRA2 antibodies are used in various scientific applications to detect and study HTRA2 in human samples. These antibodies are typically monoclonal or polyclonal and are developed in mice. They can be used in techniques such as Western Blot, Immunohistochemistry, ELISA, Immunocytochemistry, and Immunoprecipitation .