The ALDH4A1 enzyme is a 62 kDa mitochondrial protein encoded by the ALDH4A1 gene. It catalyzes the conversion of pyrroline-5-carboxylate to glutamate, a step essential for proline degradation and amino acid homeostasis . Defects in this enzyme are linked to hyperprolinemia type 2, a metabolic disorder characterized by elevated proline levels .
The ALDH4A1 antibody (e.g., Proteintech’s 11604-1-AP) is a rabbit polyclonal antibody validated for use in western blot (WB), immunohistochemistry (IHC), immunoprecipitation (IP), and enzyme-linked immunosorbent assay (ELISA) . It targets the full-length protein and exhibits reactivity with human, mouse, and rat samples.
Recent studies have identified ALDH4A1 as a key autoantigen in atherosclerosis, an inflammatory disease underlying cardiovascular events . Key findings include:
Infusion of A12 antibodies in Ldlr−/− mice reduced circulating free cholesterol and LDL, suggesting immunomodulatory benefits . These findings highlight ALDH4A1’s dual role as a therapeutic target and diagnostic marker.
Diagnostic Use:
ALDH4A1’s elevated plasma levels in atherosclerotic patients make it a candidate biomarker for early disease detection. Proteomic profiling of plaques has confirmed its altered distribution in affected tissues .
Therapeutic Potential:
Preclinical studies demonstrate that anti-ALDH4A1 antibodies modulate immune responses, inhibit plaque growth, and improve lipid profiles . This positions ALDH4A1 as a novel target for antibody-based therapies in cardiovascular disease.
ALDH4A1 antibodies have been validated for multiple research applications, with specific considerations for each technique:
Western Blotting (WB): Most commercially available ALDH4A1 antibodies are validated for WB at dilutions ranging from 1:500-1:5000, with the expected molecular weight of approximately 62 kDa . For optimal results, use fresh tissue lysates and include appropriate positive controls (e.g., liver tissue, skeletal muscle, or HepG2 cells).
Immunohistochemistry (IHC): For paraffin-embedded tissues, a dilution range of 1:50-1:500 is typically recommended . Heat-mediated antigen retrieval with EDTA buffer (pH 9.0) or citrate buffer (pH 6.0) significantly enhances signal detection .
Immunofluorescence (IF)/Immunocytochemistry (ICC): Antibodies can be used at approximately 1:250 dilution, with acetone fixation at -20°C showing excellent results for cellular localization studies .
Immunoprecipitation (IP): Use 0.5-4.0 μg of antibody for 1.0-3.0 mg of total protein lysate to effectively isolate ALDH4A1 from complex samples .
Flow Cytometry: For intracellular staining, 2% paraformaldehyde fixation followed by permeabilization and antibody incubation at approximately 1:160 dilution provides reliable detection .
ELISA: Both sandwich ELISA kits and individual antibodies are available for quantitative measurement of ALDH4A1 in serum, plasma, and cell culture supernatants, with detection ranges typically from 0.48-120 ng/ml .
Optimizing fixation for ALDH4A1 immunostaining requires consideration of its mitochondrial localization:
Paraformaldehyde fixation (2-4%) for 10-15 minutes at room temperature works well for most cell types but may compromise mitochondrial structure.
Cold methanol fixation (-20°C for 10 minutes) better preserves mitochondrial architecture and enhances accessibility to mitochondrial matrix proteins like ALDH4A1.
Acetone fixation at -20°C has shown excellent results for cellular localization studies as demonstrated with HeLa cells .
Dual fixation protocol: For challenging samples, a combination approach using 4% paraformaldehyde (10 minutes) followed by methanol permeabilization (-20°C, 5 minutes) may significantly improve signal-to-noise ratio.
Antigen retrieval: For FFPE tissues, heat-mediated antigen retrieval with EDTA buffer (pH 9.0) is critical, as it has been shown to be more effective than citrate buffer for exposing mitochondrial epitopes .
Include a mitochondrial marker (e.g., TOMM20, COX IV, or MitoTracker) in co-localization experiments to validate the specificity of ALDH4A1 staining.
ALDH4A1 antibodies serve as valuable tools for investigating atherosclerosis progression through multiple approaches:
Biomarker validation studies: Research has shown that circulating ALDH4A1 is increased in both mice and humans with atherosclerosis, making it a promising biomarker . Researchers can employ ELISA or western blotting with anti-ALDH4A1 antibodies to quantify plasma or serum ALDH4A1 levels.
Tissue distribution analysis: The distribution of ALDH4A1 is altered during atherosclerosis, with immunohistochemistry revealing changes in both medial and intimal layers of vessels . The table below summarizes findings on ALDH4A1 abundance in different vessel layers during disease progression:
| Tissue Layer | Healthy Vessels | Fatty Streak Lesions | Fibrolipid Lesions |
|---|---|---|---|
| Media | Normal expression | Altered distribution | Significantly altered |
| Intima | Minimal expression | Increased expression | Markedly increased |
Therapeutic potential evaluation: The A12 antibody (specific to ALDH4A1) has demonstrated protective effects against atherosclerosis when infused into Ldlr -/- mice, delaying plaque formation and reducing circulating free cholesterol and LDL . Researchers can design similar intervention studies using:
Anti-ALDH4A1 antibodies for in vivo treatment
Oil-Red staining of en face aortas to assess plaque formation
Lipid profile analysis following antibody administration
Flow cytometry to evaluate immune cell populations in atherosclerotic lesions
Mechanistic investigations: Employ anti-ALDH4A1 antibodies in competition assays with potential binding partners (e.g., MDA-LDL) to understand interaction mechanisms relevant to atherosclerosis pathophysiology.
Designing experiments to investigate ALDH4A1 auto-antibody responses in atherosclerosis requires careful planning:
Ldlr -/- mice fed high-fat diet (HFD) for 16 weeks represent a well-established model that demonstrates increased anti-ALDH4A1 antibody production
Consider the timeframe for antibody development (anti-ALDH4A1 IgM appears earlier than IgG)
Include appropriate controls: Ldlr +/+ mice on normal diet (ND), Ldlr +/+ on HFD, and Ldlr -/- on ND
Single-cell sorting of splenic germinal center B cells and plasma cells
PCR amplification and sequencing of IgH and IgL cDNAs
Identify expanded B cell clones in atherogenic conditions
Express representative antibodies for functional testing
ELISA assays using recombinant ALDH4A1 protein
Competition immunoassays with potential cross-reacting antigens
Immunoprecipitation followed by mass spectrometry to confirm binding specificity
Immunization protocols with ALDH4A1 plus adjuvant
Flow cytometric analysis of germinal center B cells (Fas+GL7+) and IgG1+ B cells
Quantification of ALDH4A1-specific antibody responses
Rigorous experimental design requires appropriate controls when working with ALDH4A1 antibodies:
Tissue samples with known high ALDH4A1 expression (liver, skeletal muscle, heart)
Recombinant ALDH4A1 protein for antibody specificity testing
Primary antibody omission to assess secondary antibody non-specific binding
Isotype-matched control antibodies (e.g., mGO53 for monoclonal antibodies)
Tissues or cells from ALDH4A1 knockout models (when available)
siRNA or shRNA knockdown samples for partial expression reduction
Peptide competition/neutralization assays using the immunizing peptide
Cross-adsorption experiments to test for cross-reactivity with related ALDH family members
Validation in multiple applications (e.g., if positive in WB, confirm with IHC or IP)
Technical replicates (minimum of 3)
Biological replicates from independent samples
Use of multiple antibodies targeting different epitopes of ALDH4A1
Co-staining with mitochondrial markers (essential given ALDH4A1's mitochondrial localization)
Cell fractionation followed by western blotting to confirm enrichment in mitochondrial fraction
When facing discrepancies in ALDH4A1 detection, systematic troubleshooting can help resolve inconsistencies:
Compare epitope locations - antibodies targeting different regions may yield different results
Review validation data provided by manufacturers regarding specificity and cross-reactivity
Consider antibody formats - monoclonal antibodies offer higher specificity but may be sensitive to epitope masking, while polyclonal antibodies provide broader detection but potentially more background
Fixation methods significantly impact epitope accessibility, especially for mitochondrial proteins
Antigen retrieval conditions (buffer pH, temperature, duration) may need optimization
Extraction methods may differentially preserve protein conformation or post-translational modifications
For WB vs. IHC discrepancies: Proteins may be denatured in WB but maintain native conformation in IHC
For IHC vs. IF discrepancies: Tissue processing methods and fixation protocols differ substantially
For IP vs. ELISA discrepancies: Antibody affinity may vary between liquid-phase and solid-phase binding
ALDH4A1 expression shows tissue-specific patterns with highest levels in liver, followed by skeletal muscle, kidney, heart, and other tissues
Disease states like atherosclerosis alter ALDH4A1 distribution and abundance
Consider potential post-translational modifications or splice variants
Employ orthogonal detection methods (e.g., mass spectrometry)
Use genetic approaches (CRISPR/Cas9 knockout) to validate antibody specificity
Consult RNA-seq data to correlate protein detection with transcript levels
Consider performing western blots under non-reducing conditions if epitope recognition is affected by disulfide bonds
ALDH4A1 antibodies provide valuable tools for investigating enzyme inhibition in proline metabolism disorders:
Use purified ALDH4A1 (immunoprecipitated with specific antibodies) for in vitro enzyme assays
Screen potential inhibitors (such as proline and hydroxyproline stereoisomers) for inhibitory effects
Determine inhibition constants (Ki) for competitive inhibitors like trans-4-hydroxy-L-proline (0.7 mM) and L-proline (1.9 mM)
Analyze inhibition mechanisms (competitive, non-competitive, uncompetitive) using Lineweaver-Burk plots
Employ antibodies in co-crystallization experiments to stabilize enzyme conformations
Use antibody-based pull-downs to isolate ALDH4A1-inhibitor complexes for structural analysis
X-ray crystallography has revealed that stereospecific inhibition by trans-4-hydroxy-L-proline involves serine residue hydrogen bonding to the amine group
ALDH4A1 deficiency is associated with type II hyperprolinemia
Antibodies can help characterize mutant ALDH4A1 proteins from patient samples
Use immunohistochemistry to study ALDH4A1 distribution in affected tissues
Develop assays to screen for compounds that might rescue defective ALDH4A1 function
Quantify ALDH4A1 levels in biological fluids from patients with proline metabolism disorders
Correlate enzyme levels with clinical parameters and disease severity
Analyze antibody-accessible epitopes in mutant proteins to understand structural consequences of mutations
ALDH4A1's dual role in metabolizing substrates from both proline and hydroxyproline pathways can be investigated using antibody-based approaches:
Generate conformation-specific antibodies that preferentially recognize ALDH4A1 bound to either L-glutamate-γ-semialdehyde (proline pathway) or 4-hydroxy-L-glutamate-γ-semialdehyde (hydroxyproline pathway)
Use these antibodies to track substrate-specific conformational changes in different physiological contexts
Employ antibodies in epitope mapping experiments to identify regions critical for dual substrate recognition
Develop antibodies that differentially inhibit activity toward one substrate versus the other
Structural analysis has shown that trans-4-hydroxy-L-proline is the strongest inhibitor (Ki = 0.7 mM), suggesting preferential interaction with the active site
Use antibodies to immunoprecipitate ALDH4A1 along with associated proteins from cells treated to activate either proline or hydroxyproline catabolism
Identify pathway-specific binding partners or post-translational modifications
Analyze how these interactions might regulate substrate preference in different tissues
| Technique | Application for Dual Substrate Analysis | Key Considerations |
|---|---|---|
| Co-immunoprecipitation | Identify substrate-specific binding partners | Use appropriate detergents to preserve protein-protein interactions |
| Proximity ligation assay | Visualize ALDH4A1 interactions in situ | Requires antibodies raised in different species |
| ELISA | Quantify ALDH4A1 with different substrates | May require substrate-locked enzyme conformations |
| Immunofluorescence | Localize ALDH4A1 in tissues with different metabolic demands | Co-stain with markers of proline vs. hydroxyproline metabolism |
| Hydrogen-deuterium exchange MS | Map conformational changes upon substrate binding | Use antibodies to capture specific conformational states |
Develop assays to understand how ALDH4A1 balances its dual roles in proline and hydroxyproline catabolism
Investigate potential substrate inhibition mechanisms, as research indicates that hydroxyproline catabolism may be inhibited by trans-4-hydroxy-L-proline, similar to how proline catabolism is inhibited by L-proline
ALDH4A1 shows promise as a biomarker for atherosclerosis, with antibody-based detection methods offering translational potential:
Sandwich ELISA using capture and detection antibodies targeting different ALDH4A1 epitopes
Calibrate using recombinant ALDH4A1 standards (0.48-120 ng/ml range)
Validate in cohorts of patients with atherosclerosis versus healthy controls
Assess correlation with established cardiovascular risk markers and imaging findings
Incorporate anti-ALDH4A1 antibodies into multiplex platforms to simultaneously detect multiple atherosclerosis-related biomarkers
Combine with other established markers (e.g., high-sensitivity C-reactive protein, lipoprotein-associated phospholipase A2)
Develop risk prediction algorithms incorporating ALDH4A1 levels
Research has shown circulating ALDH4A1 is increased in atherosclerosis patients
Logistic regression analysis adjusted for cardiovascular risk factors supports ALDH4A1's potential as an independent predictor
Standardize pre-analytical variables (sample collection, processing, storage)
Establish reference ranges across different demographics
Adapt antibody-based detection to lateral flow or microfluidic platforms
Optimize antibody pairs for sensitivity and specificity in complex matrices (whole blood, plasma)
Evaluate correlation with laboratory-based quantitative methods
The discovery that anti-ALDH4A1 antibodies can protect against atherosclerosis progression opens avenues for therapeutic development:
Humanize or fully human antibody development based on the protective A12 antibody identified in mouse models
Engineer antibody properties (affinity, specificity, effector functions) for optimal therapeutic effect
Evaluate various antibody formats (IgG, Fab, scFv) for tissue penetration and pharmacokinetics
Consider bispecific antibodies targeting ALDH4A1 and additional atherosclerosis-related targets
Define how anti-ALDH4A1 antibodies reduce circulating free cholesterol and LDL
Investigate effects on immune cell infiltration into atherosclerotic plaques
Determine if antibodies neutralize enzymatic activity or alter ALDH4A1 distribution
Assess impact on mitochondrial function in vascular cells
Dose-response studies in atherosclerosis models (Ldlr -/- mice)
Compare prophylactic versus therapeutic intervention timing
Evaluate safety profile (immunogenicity, off-target effects)
Develop biomarkers to monitor treatment response (circulating ALDH4A1, lipid profiles)
Patient stratification strategies (e.g., based on ALDH4A1 levels or genetic variants)
Combination approaches with standard-of-care therapies (statins, PCSK9 inhibitors)
Development of companion diagnostics to identify patients most likely to benefit
Route of administration optimization (intravenous, subcutaneous)
Immunoprecipitation of ALDH4A1 from mitochondrial preparations requires attention to several critical parameters:
Use differential centrifugation with sucrose gradient for higher purity
Assess mitochondrial fraction purity using markers (COX IV, TOMM20)
Consider tissue source carefully, as ALDH4A1 expression varies (highest in liver, skeletal muscle, kidney)
Gentle detergents (0.5-1% NP-40 or digitonin) better preserve mitochondrial protein complexes
Include protease inhibitors to prevent degradation
Maintain physiological pH (7.2-7.4) to preserve native conformation
Consider adding NAD+ (ALDH4A1 cofactor) to stabilize enzyme structure
Use affinity-purified antibodies specifically validated for IP applications
Optimal antibody amount: 0.5-4.0 μg per 1.0-3.0 mg of total protein lysate
Pre-clear lysates to reduce non-specific binding
Consider using magnetic beads conjugated with antibodies for gentler elution
Glycine elution (pH 2.5-3.0) for applications requiring native protein
SDS sample buffer for subsequent SDS-PAGE analysis
Peptide competition elution for higher specificity
Analyze enrichment of ALDH4A1 in immunoprecipitates compared to input using PSM (peptide spectrum match) ratios
Research has shown enrichment values of approximately 3-fold in successful ALDH4A1 immunoprecipitation experiments
Detecting ALDH4A1 in atherosclerotic plaques requires specific modifications to standard immunohistochemical protocols:
Rapid fixation is crucial to preserve mitochondrial antigens
Perfusion fixation (in animal models) provides superior results compared to immersion fixation
Use 4% paraformaldehyde for 24-48 hours followed by careful paraffin embedding
Consider using frozen sections for antigens particularly sensitive to processing
Heat-mediated antigen retrieval with EDTA buffer pH 9.0 has shown superior results for ALDH4A1
Extended retrieval times (20-30 minutes) may be necessary for lipid-rich plaque samples
Pressure cooker-based retrieval often provides more consistent results than water bath methods
Amplification systems (e.g., tyramide signal amplification) may be necessary for detecting low abundance ALDH4A1
Use polymer-based detection systems to reduce background in lipid-rich tissues
Consider chromogens that provide good contrast with lipid-rich plaque components (e.g., AEC or ImmPACT SG)
Extended blocking (2-3 hours) with serum-free protein block
Include lipid blocking steps (e.g., pre-treatment with absolute alcohol)
Use Sudan Black B (0.1-0.3%) to reduce autofluorescence from lipofuscin