ACLB-1 is a subunit of ATP-citrate synthase (ACLB), an enzyme essential for catalyzing the synthesis of citrate from acetyl-CoA and oxaloacetate. This reaction is pivotal in lipid biosynthesis and energy metabolism, particularly in plants such as Arabidopsis thaliana .
Species | Common Name | Reactivity Confirmed? |
---|---|---|
Arabidopsis thaliana | Thale cress | Yes |
Zea mays | Maize | Yes |
Oryza sativa | Rice | Yes |
Physcomitrium patens | Moss | Yes |
Glycine max | Soybean | Yes |
Western Blotting: Detects ACLB-1 in lysates from Arabidopsis and other plant tissues .
ELISA: Quantifies ACLB-1 expression levels under varying metabolic conditions .
Functional Studies: Used to investigate ACLB-1’s role in lipid biosynthesis and stress responses in crops like maize and rice .
Metabolic Regulation: ACLB-1 expression correlates with lipid accumulation in developing seeds of Brassica napus .
Stress Adaptation: Elevated ACLB-1 levels were observed in drought-stressed Arabidopsis, suggesting a role in metabolic adaptation .
Specificity: Targets a conserved C-terminal epitope, minimizing cross-reactivity with non-homologous proteins .
Broad Utility: Validated across diverse plant species, enabling comparative studies in plant metabolism .
Antibodies are versatile tools in protein research with applications including:
Western Blot (WB): For protein detection with typical dilutions of 1:1000-1:4000
Immunoprecipitation (IP): Using 0.5-4.0 μg for 1-3 mg of total protein lysate
Immunohistochemistry (IHC): With dilutions ranging from 1:50-1:500
Immunofluorescence (IF)/Immunocytochemistry (ICC): At dilutions of 1:10-1:100
Flow Cytometry (FC): Typically using 0.40 μg per 10^6 cells in a 100 μl suspension
For optimal results, antibody dilutions should be titrated in each testing system as sensitivity can be sample-dependent. Published literature often provides validation data for specific applications, which can serve as a starting point for new experimental designs .
Proper storage is critical for maintaining antibody activity:
Store antibodies at -20°C in appropriate buffer systems (e.g., PBS with 0.02% sodium azide and 50% glycerol at pH 7.3)
Antibodies are typically stable for one year after shipment when stored correctly
For small volume antibodies (e.g., 20μl), including 0.1% BSA can increase stability
Aliquoting is generally unnecessary for -20°C storage but may be beneficial for frequently used antibodies to avoid freeze-thaw cycles
When planning experiments, consider antibody stability under working conditions and minimize exposure to room temperature for extended periods.
Researchers should evaluate:
Tested reactivity across species (human, mouse, rat, etc.)
Molecular weight confirmation (predicted vs. observed)
Application-specific validation data (positive results in relevant models)
Knockout/knockdown validation when available
Published literature using the antibody for similar applications
For example, ACLY antibodies might show reactivity in multiple species with an observed molecular weight of approximately 120 kDa that matches the calculated weight of 121 kDa, providing confidence in antibody specificity .
When designing antibody-mediated cell activation assays:
Establish appropriate cell ratios (e.g., 1×10^4 target cells with 1×10^5 effector cells)
Block non-specific binding using Fc block (e.g., 150 ng per well)
Prepare antibody serial dilutions (e.g., 10-fold from 50 nM)
Include appropriate activation markers (CD25, CD69) and viability markers
Calculate cytotoxicity using the formula: [1-(live cell numbers in treated group/live cell numbers in control group)]×100
For bispecific antibodies like those targeting CLL-1 and CD3, assessing both target cell death and T-cell activation simultaneously provides comprehensive functional data. Flow cytometry analysis should include markers to distinguish target populations (e.g., CD33 for AML cells) and activation markers for effector cells .
Addressing cross-reactivity requires:
Comprehensive binding analysis against intended targets and structurally similar proteins
Flow cytometry assessment of binding to various cell types (e.g., granulocytes, monocytes, myeloid dendritic cells, natural killer cells, B cells)
Comparison of binding EC50 values across cell lines expressing the target at different levels
For example, when developing antibodies against targets like CLL-1, researchers should confirm binding to known CLL-1-expressing cells (EC50 values around 0.15 nM for high-expressing cell lines) while verifying minimal binding to non-expressing cells .
Designing rigorous in vivo experiments requires:
Selection of appropriate models, including both cell line-derived xenografts and patient-derived xenografts
Consideration of orthotopic models to better recapitulate the tumor microenvironment
Implementation of multiple dosing regimens to establish dose-response relationships
Comprehensive endpoint analyses including tumor volume, immunohistochemistry, and ex vivo functional assays
Assessment of mechanistic outcomes beyond tumor growth (e.g., changes in stemness, metabolism, oncogenic signaling)
Research with claudin-1-specific antibodies demonstrated the importance of using diverse model systems, with effects confirmed in both cell line-based models and patient-derived 3D ex vivo models before proceeding to in vivo validation .
Key factors affecting IHC performance include:
Antigen retrieval method: Different antibodies require specific retrieval methods (e.g., TE buffer pH 9.0 vs. citrate buffer pH 6.0)
Antibody dilution: Typically ranging from 1:50-1:500 for IHC applications
Incubation conditions: Temperature and duration significantly impact sensitivity
Detection system: Selection of appropriate secondary antibodies and visualization methods
Tissue preparation: Fixation method and duration affect epitope accessibility
When troubleshooting IHC, researchers should systematically optimize each variable while maintaining appropriate positive and negative controls. For example, ACLY antibody detection in human prostate cancer tissue requires specific antigen retrieval with TE buffer pH 9.0 .
Quantitative assessment of antibody binding requires:
Determination of EC50 values through dose-response curves
Measurement of binding kinetics (kon and koff rates) using surface plasmon resonance
Comparative binding analysis across different target-expressing cell lines
Competition assays to evaluate epitope specificity
Cross-reactivity assessment against structurally similar targets
For bispecific antibodies, researchers should evaluate binding to each target independently and assess whether binding to one target affects binding to the other. Flow cytometry analysis provides a platform to quantify binding to native targets on cell surfaces .
When working with ADCs, researchers must consider:
Conjugation chemistry: The linker choice affects stability and drug release (e.g., cleavable linkers like GGFG)
Drug-to-antibody ratio: Optimization is required for maximum efficacy with minimal off-target effects
Payload selection: Different payloads (e.g., topoisomerase I inhibitors) have distinct mechanisms of action
Internalization kinetics: Efficient internalization is crucial for cytotoxic payload delivery
Bystander effect: Some ADCs can affect neighboring cells through payload diffusion
For example, trastuzumab deruxtecan (Enhertu) employs a cleavable GGFG linker and a topoisomerase I inhibitor payload, which has specific implications for experimental design when studying HER2-targeted therapies .
Bispecific antibodies offer unique research advantages:
Simultaneous targeting of tumor cells and immune effectors without genetic modification
Format-dependent activity: 2+1 formats can show enhanced potency compared to 1+1 formats
Concentration-dependent T-cell activation specific to target-expressing cells
Potential to overcome resistance mechanisms to conventional antibody therapies
Ability to redirect endogenous T cells against cancer cells lacking typical immunogenic markers
Research with bispecific antibodies like ABL602, which targets CLL-1 and CD3, demonstrates how these molecules can activate T cells in a target-dependent manner, producing potent cytotoxicity against AML cells with EC50 values in the sub-nanomolar range .
Complex antibody effects require multifaceted analysis:
Single-cell RNA sequencing to characterize differential effects on cell subpopulations
Assessment of stemness markers in treated vs. untreated tumors
Metabolic profiling to identify changes in cancer cell energetics
Analysis of immune cell infiltration and activation state
Research with claudin-1-specific antibodies revealed that targeting non-junctional claudin-1 affected tumor growth through multiple mechanisms, including regulation of tumor stemness, metabolism, oncogenic signaling, and perturbation of the tumor immune microenvironment .
Key considerations include:
Sequence source optimization: Humanized or fully human sequences minimize immunogenicity
Backbone selection: Different IgG subtypes (IgG1, IgG2, etc.) have distinct effector functions
Fc modifications: Engineered modifications can enhance or reduce specific effector functions
Expression system selection: CHO cells vs. other systems affect glycosylation patterns
Light chain pairing: Kappa vs. lambda affects stability and manufacturing
The antibody therapeutic database demonstrates the diversity of approaches to antibody development, with various sequence sources, backbones, and expression systems selected based on the intended therapeutic application .
To minimize non-specific binding:
Optimize blocking conditions (type of blocking agent, concentration, duration)
Adjust antibody dilution (typically 1:1000-1:4000 for Western blot applications)
Increase washing stringency (buffer composition, number of washes, duration)
Consider sample preparation modifications to reduce background
Include appropriate controls (lysates from knockout/knockdown samples)
For research with ACLY antibodies, validation across multiple cell types (L02, A549, HeLa, Jurkat, K-562, MCF-7 cells) and tissues (mouse and rat liver) helps establish the pattern of specific binding and identify potential cross-reactivity .
When facing contradictory results:
Compare epitopes recognized by different antibodies used
Evaluate fixation and sample preparation effects on epitope accessibility
Consider post-translational modifications that might affect antibody recognition
Verify results with genetic approaches (siRNA, CRISPR) to confirm specificity
Research demonstrating effects of claudin-1 antibodies on tumor growth employed multiple methodologies, including 3D ex vivo models and various in vivo models, to ensure robust and reproducible findings across different experimental systems .
Comprehensive validation strategies include:
Genetic knockout/knockdown controls to confirm specificity
Peptide competition assays to verify epitope specificity
Testing across multiple applications (WB, IHC, IF) to confirm consistent recognition
Comparison of results with multiple antibodies targeting different epitopes of the same protein
Mass spectrometry confirmation of immunoprecipitated proteins
When development of therapeutic antibodies like those targeting claudin-1, extensive validation in both in vitro and in vivo systems provided strong evidence of specificity before proceeding to mechanistic studies and therapeutic development .