The DDO Antibody specifically binds to the D-aspartate oxidase protein, encoded by the DDO gene in humans. The protein is localized to peroxisomes and belongs to the DAMOX/DASOX family, with isoforms generated through alternative splicing . It serves as a key metabolic enzyme, catalyzing reactions dependent on flavin adenine dinucleotide (FAD) or 6-hydroxy-FAD .
The DDO Antibody is utilized across multiple experimental platforms to study protein expression, localization, and functional roles.
IHC-Paraffin: Strong cytoplasmic staining in exocrine glandular cells of human pancreas .
WB: Detects DDO in transfected 293T cell lysates at ~41 kDa .
Reactivity: Primarily validated for human samples, with cross-reactivity reported for mouse and rat in some formulations .
A range of DDO Antibodies is available, differing in host species, epitope specificity, and conjugation. Below is a comparison of select products:
Polyclonal vs. Monoclonal: Polyclonal antibodies (e.g., Atlas, Bio-Techne) offer broader epitope recognition, while monoclonal antibodies (e.g., Thermo Fisher) provide higher specificity .
Conjugates: Options include unconjugated, biotin, APC, and HRP-labeled variants for versatile detection .
Western Blot: Detects DDO in transfected lysates but not in non-transfected controls .
IHC-Paraffin: Requires antigen retrieval (e.g., HIER pH 6) for optimal staining .
Species Cross-Reactivity: Limited to human and rodent models in most formulations .
While direct research findings from the provided sources focus on technical applications, the DDO Antibody underpins studies investigating:
D-aspartate oxidase (DDO) is a protein encoded by the DDO gene. In humans, the canonical protein has 341 amino acid residues with a molecular mass of 37.5 kDa and is primarily localized in peroxisomes. It belongs to the DAMOX/DASOX protein family and is involved in metabolic processes. Alternative splicing produces four different isoforms of this protein .
DDO antibodies are immunological tools used for the detection and analysis of the DDO protein in various experimental contexts. These antibodies are commonly employed in techniques such as:
Western blotting for protein identification
Immunohistochemistry for tissue localization studies
ELISA for quantitative analysis
Immunocytochemistry for cellular localization
Researchers use these antibodies to investigate DDO expression patterns, subcellular localization, and functional roles in different physiological and pathological conditions .
DDO268 is a synthetic virus-derived oligonucleotide developed for use as a vaccine adjuvant. It is derived from the 546-nucleotide-long Sendai virus nonstandard viral genome, which serves as a primary immunostimulatory molecule during infections .
As an adjuvant, DDO268 functions through the following mechanisms:
When co-packaged with mRNA in lipid nanoparticles, it activates RIG I-like receptors and TLR3
It safely induces local type I interferon (IFN) production at the site of inoculation without systemic effects
It stimulates dendritic cell type 1 (DC1) activation and migration to draining lymph nodes
It improves the generation of IgG2c antibodies and antigen-specific Th1 CD4+ and CD8+ T-cells (IFNγ+TNFα+IL2+)
This adjuvant activity makes DDO268 particularly valuable for mRNA vaccines targeting conserved viral epitopes, as demonstrated in influenza A virus (IAV) nucleoprotein mRNA vaccine research .
DDO268 enhances both humoral and cellular immune responses in mRNA vaccine platforms through several interconnected mechanisms:
RIG-I pathway activation: When delivered intracellularly via lipid nanoparticles, DDO268 activates RIG-I-like receptors, which are expressed in most nucleated cells. This near-universal expression increases the likelihood that DDO268's adjuvant activity will be conserved across experimental models and in humans .
Localized immune stimulation: DDO268 induces a localized immune response at the injection site without detectable systemic effects, creating a controlled inflammatory environment that promotes antigen processing and presentation .
Dendritic cell activation: The adjuvant triggers dendritic cell type 1 activation and migration to draining lymph nodes, enhancing antigen presentation to T cells .
Enhanced T-cell responses: DDO268 significantly improves the generation of antigen-specific Th1 CD4+ and CD8+ T-cells that express multiple cytokines (IFNγ+TNFα+IL2+), creating a more robust cellular immune response .
Antibody production: The adjuvant enhances the generation of IgG2c antibodies, contributing to humoral immunity .
Dose-sparing effect: Notably, the inclusion of DDO268 reduces the antigen dose required to achieve protection, making it potentially more cost-effective for vaccine development .
Recent studies with influenza A virus nucleoprotein mRNA vaccines demonstrate that these enhanced immune responses translate to improved protection against lethal viral challenge in mouse models .
Detection and characterization of anti-drug antibodies (ADAs) against DDO-containing therapeutics requires robust methodological approaches:
Recommended methods for ADA detection:
Immunoassay-based approaches:
Enzyme-linked immunosorbent assays (ELISAs): The standard approach involves immobilizing the drug on a solid surface, adding patient serum, and detecting bound antibodies with labeled anti-human antibodies .
Homogeneous mobility shift assays: These solution-phase assays detect ADAs without separation steps, minimizing false positives due to solid-phase binding artifacts .
Advanced analytical techniques:
Surface plasmon resonance (SPR) spectroscopy: Provides real-time, label-free detection of ADAs with information on binding kinetics and affinity .
Capillary electrophoresis: Useful for separating and characterizing ADAs based on size, charge, and binding properties .
Liquid chromatography-mass spectrometry (LC-MS): Enables precise identification and characterization of ADAs at the molecular level .
Functional assays:
Characterization strategies:
Isotype profiling to determine ADA class (IgG, IgM, IgE)
Epitope mapping to identify binding regions
Affinity determination using SPR or other binding assays
Neutralization assessment to determine functional impact
When designing ADA detection protocols, researchers should implement drug tolerance steps to minimize drug interference and include appropriate positive and negative controls to ensure assay validity .
DDO268 represents a distinct approach to adjuvanting mRNA vaccines compared to other adjuvants through its RIG-I pathway activation:
Comparative analysis of adjuvant mechanisms:
Adjuvant Type | Primary Receptors | Immune Response Profile | Safety Profile | Application in mRNA Vaccines |
---|---|---|---|---|
DDO268 | RIG-I-like receptors and TLR3 | Type I IFN production, DC1 activation, Th1-biased CD4+/CD8+ responses | Localized immune stimulation without detectable systemic effects | Co-packaged with mRNA in LNPs |
TLR agonists (e.g., CpG) | TLR9 | Pro-inflammatory cytokines, B-cell activation, DC maturation | Potential systemic inflammation | Often administered separately |
Saponin-based adjuvants | Membrane interactions | Balanced Th1/Th2 responses, strong antibody production | Hemolytic potential at high doses | Challenging to incorporate with LNPs |
Alum | NLRP3 inflammasome | Th2-biased, strong antibody responses | Established safety record | Not typically used with mRNA platforms |
Unique advantages of DDO268:
Universal expression of target receptors: RIG-I is expressed in most nucleated cells, increasing the likelihood that DDO268's adjuvant activity will be conserved across experimental models and humans .
Compatibility with LNP delivery: DDO268 can be co-packaged with mRNA in lipid nanoparticles, ensuring simultaneous delivery of adjuvant and antigen to the same cells .
Balanced immune stimulation: DDO268 induces both humoral (IgG2c antibodies) and cellular (CD8+ T-cells) immune responses, addressing a key challenge in viral vaccine development .
Dose-sparing effect: The inclusion of DDO268 reduces the antigen dose required to achieve protection, potentially improving cost-effectiveness .
Safety profile: DDO268 induces localized immune responses without detectable systemic effects, as demonstrated by studies showing no significant changes in blood parameters, serum chemistry, or systemic cytokine levels after administration .
The research indicates that DDO268's ability to safely trigger RIG-I-dependent signaling provides advantages for mRNA vaccines targeting conserved viral epitopes, particularly when strong cellular immunity is desired .
When designing experiments using DDO antibodies for detecting D-aspartate oxidase, researchers should address several critical considerations:
Pre-experimental planning:
Antibody validation strategy:
Cross-reactivity assessment: Confirm specificity for DDO versus other DAMOX/DASOX family members
Validation across multiple detection methods (Western blot, IHC, ELISA)
Positive and negative control tissues/cell lines expressing different levels of DDO
Isotype and conjugate selection:
Species reactivity considerations:
Methodology optimization:
Western blot protocol refinement:
Optimize protein extraction for peroxisomal proteins
Determine optimal antibody concentration through titration experiments
Consider detection method sensitivity requirements (chemiluminescence vs. fluorescence)
Immunohistochemistry/immunocytochemistry considerations:
Optimize fixation methods for peroxisomal protein preservation
Evaluate antigen retrieval requirements
Implement proper blocking to reduce background
Consider multiplexing to co-localize with peroxisomal markers
Addressing alternative splicing:
Quantitative considerations:
For quantitative applications, establish standard curves
Implement appropriate normalization controls
Consider dynamic range limitations
Technical controls:
Include isotype controls to assess non-specific binding
Implement blocking peptide controls when available
Consider genetic knockdown/knockout validation where feasible
By systematically addressing these considerations, researchers can design more robust experiments that yield reliable and reproducible results when using DDO antibodies.
Optimizing DDO268 dosage in mRNA vaccine formulations requires a systematic approach balancing immunogenicity and safety:
Dose optimization strategy:
Dose-response assessment:
Conduct dose-escalation studies (e.g., 5, 10, 25, 50 μg) to determine the minimum effective dose
Evaluate both immune response parameters and safety indicators at each dose level
Research indicates that even 10× higher doses (50 μg) than typically used can be safe, suggesting a wide therapeutic window
Antigen-adjuvant ratio optimization:
Delivery system considerations:
Lipid nanoparticle (LNP) composition affects both transfection efficiency and adjuvant activity
Optimize co-packaging of DDO268 with mRNA in LNPs to ensure simultaneous delivery
Evaluate particle size, charge, and stability of different formulations
Safety monitoring parameters:
Localized vs. systemic effects:
Temporal analysis:
Efficacy assessment:
Immune response evaluation:
Protection studies:
Challenge studies with appropriate pathogen models (e.g., influenza)
Compare protection levels across different dose formulations
Evaluate both immediate protection and durability of immune responses
In published research, DDO268 demonstrated safety even at doses 10 times higher than typically used (50 μg), with no significant changes in blood parameters, serum chemistry, or systemic cytokine levels, suggesting a favorable safety profile that facilitates dose optimization .
Understanding the structural characteristics of antibodies targeting DDO or DDO268-containing complexes requires sophisticated methodological approaches:
Computational structural prediction methods:
RosettaAntibody and related tools:
RosettaAntibody predicts 3D structure of antibodies from sequence
Uses canonical loop conformations from experimental structures
Performs energetic calculations to minimize loops
Applies docking methodology to refine VL-VH orientation
Enables de novo prediction of complementarity determining region (CDR) H3 loop
RosettaAntibodyDesign (RAbD):
Enables both de novo antibody design and affinity maturation
Classifies the antibody into regions (framework, canonical loops, HCDR3 loop)
Allows for sequence and graft design based on canonical clusters
Utilizes cluster-based CDR dihedral constraints
SnugDock methodology:
Experimental structure determination approaches:
X-ray crystallography:
Provides high-resolution structures of antibody-antigen complexes
Requires crystallization of the complex
May be challenging for flexible complexes or membrane-bound antigens
Cryo-electron microscopy (Cryo-EM):
Enables visualization of antibody-antigen complexes in their native state
Does not require crystallization
Particularly valuable for larger complexes like antibody-LNP interactions
Hydrogen-deuterium exchange mass spectrometry (HDX-MS):
Maps binding interfaces and conformational changes
Provides information on dynamics not captured by static structural methods
Useful for epitope mapping of antibody-antigen interactions
Functional analysis to complement structural studies:
Surface plasmon resonance (SPR):
Epitope mapping techniques:
Peptide arrays to identify linear epitopes
Mutagenesis studies to confirm structurally important residues
Competition assays to determine epitope overlap
For computationally designed antibodies, validation typically requires approximately 1,000 CPU-hours for antibody modeling and 250 CPU-hours for antibody-antigen docking, highlighting the computational intensity of these approaches .
DDO268 adjuvant technology shows significant potential for application across various mRNA vaccine platforms beyond influenza:
Potential applications in other viral vaccines:
Coronaviruses (including SARS-CoV-2):
DDO268's ability to enhance T-cell responses against conserved viral proteins could be valuable for coronavirus vaccines targeting conserved epitopes beyond the spike protein
The technology could potentially address the challenge of variant escape by focusing immune responses on more conserved viral components
Machine learning approaches being developed for antibody design against coronaviruses could be enhanced by incorporating DDO268 adjuvant strategies
Respiratory syncytial virus (RSV):
RSV vaccines face challenges in generating balanced humoral and cellular immunity
DDO268's demonstrated ability to enhance both antibody and T-cell responses makes it a promising candidate for RSV mRNA vaccines
HIV vaccines:
HIV's high mutation rate makes conserved epitope targeting crucial
DDO268's enhancement of CD8+ T-cell responses could benefit HIV vaccine approaches focusing on conserved regions
Implementation strategies:
Combination with other adjuvant technologies:
DDO268 could be used in complementary combinations with other adjuvants targeting different immune pathways
Synergistic effects might be achieved through strategic co-formulation
Incorporation into multi-antigen platforms:
DDO268 could enhance efficacy of mRNA vaccines delivering multiple antigens simultaneously
Particularly valuable for complex pathogens requiring broad immune responses
Personalized vaccine approaches:
Practical considerations for cross-platform application:
Antigen-specific optimization:
Different viral antigens may require adjustment of DDO268:mRNA ratios
Optimization should consider antigen size, stability, and intrinsic immunogenicity
Regulatory pathway planning:
The RIG-I pathway stimulation by DDO268 provides a mechanistic advantage that is likely to be broadly applicable across various viral vaccine platforms, as RIG-I is expressed in most nucleated cells across species , suggesting its adjuvant effects would translate well to diverse vaccine candidates.
For researchers working with DDO antibodies, establishing critical quality attributes (CQAs) and appropriate analytical methods is essential for experimental reproducibility and data reliability:
Critical quality attributes for DDO antibodies:
Specificity:
Sensitivity:
Limit of detection in various applications (Western blot, ELISA, IHC)
Dynamic range for quantitative applications
Signal-to-noise ratio under standardized conditions
Binding characteristics:
Epitope specificity (which domain/region of DDO is recognized)
Affinity/avidity parameters (KD values)
pH and buffer composition effects on binding
Functionality:
Analytical methods for characterization:
Quality control considerations:
Lot-to-lot consistency:
Implement standardized testing protocols for each production lot
Maintain reference standards for comparative analysis
Document acceptance criteria for release testing
Storage stability assessment:
Evaluate performance after various storage conditions
Establish recommended storage parameters
Define shelf-life based on retained activity
Documentation requirements:
Detailed production methods and purification steps
Comprehensive characterization data
Application-specific validation data
By establishing these critical quality attributes and implementing appropriate analytical methods, researchers can ensure reliable and reproducible results when working with DDO antibodies across different experimental contexts.
DDO268 represents a distinct class of RNA-based adjuvants with specific molecular mechanisms for stimulating innate immunity:
Comparative molecular mechanisms of RNA-based adjuvants:
DDO268's unique mechanistic features:
Dual receptor engagement:
Safety through structure:
Despite being virus-derived, DDO268 is synthetic and replication-incompetent
Its structure allows for immune activation without the risks associated with replicating viral components
Studies show no significant changes in blood parameters, serum chemistry, or systemic cytokine levels even at doses 10× higher than typically used
Cellular targeting precision:
When co-packaged with mRNA in LNPs, DDO268 ensures that cells receiving the antigen mRNA also receive the adjuvant signal
This co-localization likely contributes to its efficacy in enhancing antigen-specific responses
Downstream signaling effects:
Evolutionary advantage:
The scientific evidence indicates that DDO268's mechanism of action through RIG-I pathway activation offers advantages over other RNA-based adjuvants, particularly in terms of its safety profile and balanced stimulation of both humoral and cellular immunity .
When working with DDO antibodies, researchers may encounter several technical challenges that can affect experimental outcomes. Here are the most common issues and recommended solutions:
Potential causes:
Insufficient blocking
Excessive antibody concentration
Cross-reactivity with other DAMOX/DASOX family proteins
Solutions:
Optimize blocking conditions (try different blockers like BSA, casein, or commercial blocking solutions)
Perform careful antibody titration experiments to determine optimal concentration
Pre-adsorb antibody with related proteins when possible
Include competitive inhibition controls with recombinant DDO protein
For Western blots, increase wash stringency and duration
Potential causes:
Antibody epitope may be absent in some isoforms
Sample preparation methods may affect isoform stability
Solutions:
Verify which isoforms your antibody recognizes
Use positive controls expressing specific isoforms
Consider using multiple antibodies targeting different epitopes
Optimize protein extraction methods to preserve all isoforms
For Western blots, use gradient gels to better resolve similar-sized isoforms
Potential causes:
DDO is localized in peroxisomes, which can be challenging to preserve during fixation
Peroxisomal proteins may require specific permeabilization conditions
Antibody may not access peroxisomal compartments effectively
Solutions:
Test multiple fixation methods (PFA, methanol, acetone)
Optimize permeabilization conditions (Triton X-100, saponin, digitonin)
Co-stain with established peroxisomal markers as positive controls
Consider epitope retrieval methods if fixation affects epitope accessibility
Use confocal microscopy for better resolution of peroxisomal structures
Potential causes:
Inconsistent sample preparation
Variable transfer efficiency
Degradation of DDO protein during extraction
Solutions:
Standardize lysis buffers and include protease inhibitors
Use fresh samples when possible or implement controlled freeze-thaw procedures
Optimize transfer conditions for proteins in the ~37.5 kDa range
Consider semi-dry vs. wet transfer methods based on protein characteristics
Implement loading controls appropriate for peroxisomal proteins
Potential causes:
Limited availability of purified DDO standards
Matrix effects from complex samples
Variability in antibody lot performance
Solutions:
Develop or obtain recombinant DDO for standard curves
Perform spike-recovery experiments to assess matrix effects
Consider sandwich ELISA formats with two different antibodies
Implement quality control samples across plates and experiments
Validate each new antibody lot against a reference standard
Potential causes:
Poor antibody binding to native DDO
Inefficient antibody-bead conjugation
Co-precipitating proteins affecting specificity
Solutions:
Test different antibody-bead conjugation methods
Adjust lysis conditions to preserve native protein structure
Try cross-linking antibodies to beads to prevent antibody contamination
Consider using magnetic beads for gentler precipitation
Validate results with Western blot confirmation
By anticipating these challenges and implementing the recommended solutions, researchers can improve the reliability and reproducibility of experiments utilizing DDO antibodies.
When designing experiments to evaluate DDO268 efficacy in novel mRNA vaccine applications, researchers should consider these critical factors:
Study design framework:
Comprehensive control groups:
mRNA vaccine without adjuvant
mRNA vaccine with established adjuvants for comparison
DDO268 alone (to assess adjuvant-only effects)
Vehicle control (lipid nanoparticles without mRNA or adjuvant)
Positive control (established vaccine when available)
Dose-response evaluation:
Test multiple DDO268 concentrations (e.g., 5, 10, 25, 50 μg)
Evaluate antigen dose-sparing by testing reduced antigen doses with fixed DDO268
Assess different DDO268:mRNA ratios to identify optimal formulation
Temporal assessment points:
Early innate response (6-24 hours): Type I IFN production, dendritic cell activation
Intermediate response (7-14 days): Antibody development, T-cell priming
Peak immunity (28-35 days): Maximum antibody titers, mature T-cell responses
Durability (3-6+ months): Persistence of immunity, memory cell formation
Formulation parameters:
Lipid nanoparticle optimization:
Evaluate different LNP compositions for co-delivery of mRNA and DDO268
Assess particle size, charge, and stability
Determine encapsulation efficiency for both mRNA and DDO268
Measure in vitro transfection efficiency of various formulations
Storage stability assessment:
Test immunogenicity after various storage conditions
Evaluate freeze-thaw stability
Determine shelf-life under refrigeration and frozen conditions
Immunological assessment:
Comprehensive immune profiling:
Humoral immunity: Antibody titers, isotype distribution (focus on IgG2c), neutralization capacity
Cellular immunity: Frequency and functionality of antigen-specific CD8+ T cells (measure IFNγ+TNFα+IL2+ polyfunctional T cells)
Innate responses: Dendritic cell activation and migration, local cytokine production
Safety parameters: Local reactions, systemic biomarkers, histopathology
Mechanistic investigations:
RIG-I pathway activation confirmation using knockout models or inhibitors
Type I IFN dependency studies
Cross-presentation assessment for CD8+ T-cell priming
Antigen biodistribution and persistence studies
Protection evaluation:
Challenge models:
Homologous challenge (same strain as vaccine antigen)
Heterologous challenge (related but different strain)
Dose-response in challenge (lethal vs. sub-lethal)
Assessment of viral loads, pathology, and clinical outcomes
Correlates of protection analysis:
Identify immune parameters that correlate with protection
Develop predictive models for vaccine efficacy
Compare correlates with established vaccines when possible
Translational considerations:
Cross-species validation:
In vitro human cell studies to confirm RIG-I activation in human cells
Consider humanized mouse models for improved translation
Design with eventual GLP toxicology studies in mind
Scalability assessment:
Evaluate batch-to-batch consistency
Consider manufacturing process constraints
Implement quality control measures applicable to GMP production
Research with IAV nucleoprotein mRNA vaccines has demonstrated that DDO268 enhances both antibody and T-cell responses, providing improved protection against lethal challenge . These established protocols provide a valuable blueprint for testing DDO268 efficacy in novel mRNA vaccine applications.
Validating DDO antibody specificity and ensuring experimental reproducibility requires a systematic, multi-method approach:
Comprehensive specificity validation:
Genetic controls:
Test antibody in DDO knockout/knockdown models
Perform antibody testing in cells with DDO overexpression
Use CRISPR-edited cell lines with tagged endogenous DDO
Cross-reactivity assessment:
Multiple detection methods concordance:
Technical validation parameters:
Antibody characterization documentation:
Record complete antibody information (clone, lot, host species, isotype)
Document immunogen sequence and production method
Maintain reference standards for lot-to-lot comparison
Application-specific validation:
For Western blotting:
Determine linear dynamic range
Establish optimal antibody concentration
Verify appropriate loading controls
Document complete protocol including blocking reagents and wash conditions
For immunohistochemistry:
Optimize fixation and antigen retrieval methods
Determine optimal antibody dilution and incubation conditions
Include positive and negative tissue controls
Verify staining pattern with multiple antibodies when possible
For ELISA:
Establish standard curves with recombinant protein
Perform spike-recovery experiments in relevant matrices
Determine coefficients of variation (intra- and inter-assay)
Document detection limits and working range
Reproducibility enhancement measures:
Create detailed standard operating procedures (SOPs)
Implement quality control samples across experiments
Use automated systems where possible to reduce operator variation
Maintain consistent reagent sources and preparation methods
Data reporting and validation:
Statistical validation approach:
Pre-determine sample sizes based on power calculations
Apply appropriate statistical tests for data analysis
Report both positive and negative results
Include technical and biological replicates
Transparent reporting:
Document all antibody validation data
Report all experimental conditions in detail
Include raw data and analysis methods
Share validation protocols with collaborators or in publications
Independent verification:
Confirm key findings with alternative antibodies
Validate critical results with orthogonal methods
Consider multi-laboratory validation for key findings