ACACA (Ab-79) Antibody is a polyclonal antibody raised in rabbits that specifically recognizes the region around amino acids 78-82 (S-M-S-G-L) of human Acetyl-CoA Carboxylase alpha (ACACA) . This antibody is designed to detect both the total and phosphorylated forms of ACACA, with particular significance in recognizing the serine-79 phosphorylation site . The antibody serves as an important research tool for investigating ACACA function, regulation, and its role in fatty acid metabolism .
The antibody is commercially available from several suppliers under different catalog numbers (CSB-PA436513, PACO21567, A52041) but with consistent specifications regarding its target epitope and production method . It is typically supplied in a liquid form at a concentration of 1.0 mg/mL in a stabilizing buffer solution .
The target of this antibody, ACACA (Acetyl-CoA Carboxylase alpha), is a critical metabolic enzyme that catalyzes the rate-limiting step in the biogenesis of long-chain fatty acids . It functions as a multifunctional enzyme system carrying out three distinct functions: biotin carboxyl carrier protein, biotin carboxylase, and carboxyltransferase . ACACA is also known by several alternative names including ACC, ACAC, ACC1, ACCA, and ACACAD .
The antibody is produced through a standardized immunization protocol whereby rabbits are immunized with a synthetic peptide corresponding to amino acids 78-82 of human ACACA conjugated to keyhole limpet hemocyanin (KLH) . Following immunization, antibodies are harvested from rabbit serum and purified using affinity chromatography with the epitope-specific peptide to ensure high specificity and minimal cross-reactivity .
ACACA (Ab-79) Antibody has been validated for multiple research applications, with Western blotting and ELISA being the primary recommended uses. Table 2 outlines the recommended applications and dilutions based on manufacturer specifications.
| Application | Recommended Dilution | Notes |
|---|---|---|
| Western Blotting (WB) | 1:500-1:1000 | Detects endogenous levels of total ACACA |
| ELISA | 1:2000-1:10000 | High sensitivity for quantitative analysis |
For Western blot applications, the antibody has been shown to effectively detect ACACA in human cell lysates, producing a specific band at approximately 280 kDa . The antibody recognizes endogenous levels of total ACACA protein and can be used to study ACACA expression in various tissues and cell lines . Western blot analysis has been performed successfully with extracts from human cell lines including HeLa and HUVEC cells .
A significant aspect of the ACACA (Ab-79) Antibody is its ability to recognize the region containing the serine-79 phosphorylation site of ACACA. This site is particularly important as it represents a key regulatory mechanism for the enzyme's activity.
The phosphorylation of ACACA at serine-79 is a critical post-translational modification that regulates the enzyme's activity. Research has shown that AMP-activated protein kinase (AMPK) phosphorylates ACACA on serine-79, leading to inhibition of ACACA enzymatic activity . This regulatory mechanism plays a crucial role in cellular energy homeostasis and fatty acid metabolism.
Research findings have revealed that the serine-79 phosphorylated form of ACACA (phospho-ACACA Ser79) undergoes specific cellular relocalization during mitosis . When cells enter mitosis and the nuclear envelope breaks down, phospho-ACACA Ser79 is relocated to centrosomes, suggesting a potential non-metabolic role for this modified form of the enzyme during cell division .
Recent research utilizing ACACA (Ab-79) Antibody and other tools for studying phospho-ACACA Ser79 has revealed interesting insights into the cellular dynamics of this protein during various cellular states.
One of the most significant findings regarding phospho-ACACA Ser79 involves its behavior during mitosis. Studies have shown that the mitosis-related enhancement of phospho-ACACA Ser79 is attenuated in the presence of compound C, an AMPK inhibitor, suggesting that AMPK phosphorylates ACACA when cells enter mitosis .
Further research has demonstrated a connection between Polo-like kinase 1 (PLK1) activity and the mitotic phosphorylation of ACACA. Treatment with GW843682X, a PLK1 inhibitor, was found to abolish centrosomal activation of phospho-ACACA Ser79 during early mitosis, mimicking the causal link between PLK1 activity and the mitotic phosphorylation of AMPK's α-catalytic subunit . This finding suggests a regulatory pathway involving PLK1, AMPK, and ACACA during mitosis.
Understanding the target protein of ACACA (Ab-79) Antibody provides important context for the antibody's applications in research.
ACACA catalyzes the carboxylation of acetyl-CoA to malonyl-CoA, which represents the rate-limiting step in fatty acid synthesis . This reaction is critical for lipid metabolism and energy homeostasis in the cell. ACACA is part of a multifunctional enzyme system that carries out three distinct functions:
Biotin carboxyl carrier protein - serves as the carrier for the biotin prosthetic group
Biotin carboxylase - catalyzes the ATP-dependent carboxylation of biotin
Carboxyltransferase - transfers the carboxyl group from carboxybiotin to acetyl-CoA
While the enzymatic activity of ACACA is influenced by substrate supply and allosteric ligands, its primary short-term regulatory mechanism involves the phosphorylation of multiple serine residues by various protein kinases . Among these, the phosphorylation of serine-79 by AMPK is particularly important for inhibiting ACACA activity in response to cellular energy status .
Several antibodies targeting different epitopes of ACACA are available for research purposes. While the ACACA (Ab-79) Antibody specifically recognizes the region around amino acids 78-82, other antibodies target different regions of the protein.
Acetyl-CoA carboxylase 1 (ACC1) is a cytosolic enzyme that catalyzes the carboxylation of acetyl-CoA to malonyl-CoA. This reaction represents the initial and rate-limiting step in de novo fatty acid biosynthesis. The process proceeds in two steps: first, an ATP-dependent carboxylation of biotin carried by the biotin carboxyl carrier (BCC) domain, followed by the transfer of the carboxyl group from carboxylated biotin to acetyl-CoA.
Further research highlights the multifaceted roles of ACC1:
ACACA (Ab-79) Antibody specifically recognizes Acetyl-CoA Carboxylase, a crucial enzyme in fatty acid metabolism. There are two main types available: phospho-specific antibodies that detect ACACA only when phosphorylated at Serine 79, and total ACACA antibodies that recognize the protein regardless of phosphorylation status. The phospho-specific monoclonal antibody (such as Clone#RM270) is designed to detect human Acetyl-CoA Carboxylase specifically when phosphorylated at Ser79 and shows no cross-reactivity with the unphosphorylated form . The polyclonal variant targets a peptide sequence around amino acids 78-82 (S-M-S-G-L) derived from human Acetyl-CoA Carboxylase . This region is significant because Ser79 phosphorylation by AMPK inhibits ACC1 activity, making it an important regulatory site for studying metabolic regulation.
ACACA (Ab-79) antibodies have been validated for multiple research applications, with different variants showing specific performance characteristics:
| Antibody Type | Validated Applications | Host | Clonality | Reactive Species |
|---|---|---|---|---|
| Anti-Phospho ACC (S79) Clone#RM270 | IHC, WB | Rabbit | Monoclonal | Human |
| Acetyl-CoA Carboxylase (Ab-79) | WB | Rabbit | Polyclonal | Human |
For Western blot applications, these antibodies detect a protein of approximately 280kD (SDS-PAGE MW) . When designing experiments, researchers should consider that validation has been conducted primarily in human samples. The application scope determines experimental design parameters including sample preparation methods, blocking reagents, and detection systems appropriate for the specific technique.
For optimal activity retention, store ACACA (Ab-79) antibodies at -20°C . The monoclonal antibody is typically supplied in 50% Glycerol/PBS with 1% BSA and 0.09% sodium azide to maintain stability during storage . The polyclonal variant is generally formulated in phosphate buffered saline without Mg²⁺ and Ca²⁺ (pH 7.4, 150mM NaCl) containing 0.02% sodium azide and 50% glycerol . To preserve antibody functionality:
Aliquot upon first thaw to minimize freeze-thaw cycles
Avoid repeated freeze-thaw cycles as they can significantly reduce antibody activity
For working solutions, store at 4°C for up to one week
When retrieving from storage, thaw on ice rather than at room temperature
Centrifuge briefly after thawing to collect all material at the bottom of the tube
Following these guidelines will ensure consistent antibody performance across experiments and maximize the one-year shelf life typically specified by manufacturers .
Monoclonal and polyclonal ACACA antibodies differ in several key aspects that impact experimental design and interpretation:
When designing experiments studying ACACA phosphorylation dynamics, monoclonal antibodies provide greater specificity for monitoring the phosphorylation state at Ser79. For total protein expression studies or applications requiring detection of multiple epitopes, polyclonal antibodies may offer advantages. The choice between these antibody types should align with your specific research question and experimental needs.
Rigorous validation of ACACA (Ab-79) antibody specificity in your specific experimental system is crucial for reliable results. Implement the following comprehensive validation strategy:
Phosphatase treatment control: Split your samples and treat half with lambda phosphatase before immunoblotting with phospho-specific ACACA (Ab-79) antibody. The signal should diminish or disappear in treated samples while remaining in untreated controls.
AMPK activation/inhibition: Treat cells with AMPK activators (e.g., AICAR, metformin) or inhibitors (e.g., Compound C) to modulate ACACA phosphorylation status. The phospho-ACACA (Ser79) signal should increase with AMPK activation and decrease with inhibition.
Knockdown/knockout validation: Use siRNA-mediated knockdown or CRISPR/Cas9 knockout of ACACA to confirm antibody specificity. The signal should be reduced proportionally to knockdown efficiency or eliminated in knockout samples.
Peptide competition assay: Pre-incubate the antibody with the immunizing phosphopeptide before applying to your samples. A specific antibody will show diminished or absent signal when the detecting epitope is blocked by the competing peptide.
Cross-species reactivity assessment: Although the antibody is specified for human ACACA , if working with other species, validate cross-reactivity by comparing signals between human and target species samples under identical conditions, particularly if the epitope sequence is conserved.
Phosphorylation-specific controls: For phospho-specific antibodies, include samples from conditions known to alter ACACA phosphorylation status (e.g., starvation, insulin stimulation) as physiological controls.
Dual antibody approach: Compare results using both phospho-specific and total ACACA antibodies on the same samples to establish the ratio of phosphorylated to total protein.
Document all validation results thoroughly to establish confidence in your antibody's performance in your specific experimental system.
When encountering inconsistent results with ACACA (Ab-79) antibody, systematically evaluate these key factors:
Sample preparation protocol:
Ensure rapid sample collection and immediate denaturation to preserve phosphorylation status
Include phosphatase inhibitors (e.g., sodium fluoride, sodium orthovanadate) in lysis buffers
Standardize protein extraction methods across experiments
Verify protein concentration measurement consistency
Antibody dilution optimization:
Perform a dilution series (1:500 to 1:5000) to identify optimal signal-to-noise ratio
For Western blots, optimize both primary and secondary antibody concentrations
Document the optimal dilution for each new lot of antibody
Blocking and washing stringency:
Compare different blocking agents (BSA vs. non-fat dry milk)
Note that milk contains phosphatases that may interfere with phospho-specific detection
Increase washing stringency (more washes, higher detergent concentration) to reduce background
Detection system sensitivity:
For low abundance targets, consider using enhanced chemiluminescence substrates or fluorescent detection
Compare results using different detection methods to identify optimal sensitivity threshold
Antibody cross-reactivity assessment:
Evaluate potential cross-reactivity with ACC2 (the other ACC isoform)
Perform immunoprecipitation followed by mass spectrometry to verify antibody targets
Protein loading controls:
Include phosphorylation-insensitive controls (total protein stains)
Verify equal protein loading across all lanes
Antibody storage and handling:
Positive and negative controls:
Include lysates from cells treated with AMPK activators (positive control)
Include samples treated with phosphatases (negative control)
Creating a troubleshooting decision tree based on these factors can systematically guide resolution of inconsistent results.
Interpreting phosphorylation status data requires nuanced analysis beyond simple signal detection. Consider these methodological approaches:
Normalization strategy selection:
Always normalize phospho-ACACA (Ser79) signal to total ACACA to account for variations in total protein expression
Avoid normalizing to housekeeping proteins alone, as they don't account for changes in total ACACA levels
Consider dual normalization: first to total ACACA, then to loading controls
Signal quantification methods:
Use densitometry software with defined background subtraction protocols
Apply multiple measurement methods (peak area vs. intensity) to confirm quantification
Report phospho-to-total ACACA ratios rather than absolute phospho-signal values
Biological context interpretation:
Remember that Ser79 phosphorylation by AMPK inhibits ACC1 activity
Increased phospho-ACACA typically correlates with decreased fatty acid synthesis
Consider parallel measurement of downstream metabolites (malonyl-CoA, fatty acids) to confirm biological activity
Temporal dynamics consideration:
ACACA phosphorylation can change rapidly (minutes) in response to metabolic signals
Document precise timing of sample collection relative to treatments
Consider time-course experiments to capture phosphorylation dynamics
Physiological state influence:
Interpret results in context of feeding/fasting status, energy state, or stress conditions
Account for cell confluence and growth phase when using cultured cells
Consider that basal phosphorylation levels vary by tissue and cell type
Signal detection threshold determination:
Establish the linear range of detection for your system
Identify the lower limit of detection through dilution series
Be cautious interpreting small changes (<20%) in phosphorylation status
Multi-site phosphorylation integration:
Consider that ACACA is regulated by phosphorylation at multiple sites (not just Ser79)
Interpret Ser79 phosphorylation in context of other modifications when possible
A comprehensive interpretation requires integration of phosphorylation data with appropriate controls and contextual biological information.
When investigating the AMPK-ACC regulatory pathway, several methodological considerations are critical for generating reliable data:
Sample preparation timing:
AMPK-mediated phosphorylation of ACACA at Ser79 occurs rapidly (within minutes)
Use rapid sample collection methods (e.g., direct lysis in SDS buffer) to preserve phosphorylation state
Document precise timing between treatments and sample collection
Physiological activator selection:
Choose AMPK activators based on research question: AMP-mimetics (AICAR), biguanides (metformin), or energy stress inducers (glucose deprivation)
Include both acute and chronic activation timepoints to distinguish immediate signaling from adaptive responses
Consider dose-response curves to identify optimal activation conditions
Pathway validation approach:
Monitor multiple nodes in the pathway simultaneously (AMPK-Thr172 phosphorylation, ACC-Ser79 phosphorylation)
Include measurement of upstream activators (LKB1, CaMKK) and downstream effects (malonyl-CoA levels)
Apply AMPK inhibitors (Compound C) or activators (A-769662) as pathway controls
Isoform-specific analysis:
Consider that ACACA/ACC1 (cytosolic) and ACACB/ACC2 (mitochondrial) have different cellular localizations and functions
Design experiments to distinguish between isoforms through subcellular fractionation
Verify antibody specificity for ACC1 vs. ACC2 in your experimental system
Cell type and tissue considerations:
AMPK-ACC pathway activity varies significantly between tissues (liver, muscle, adipose)
Document cell type-specific baseline phosphorylation levels
Consider tissue-specific pathway interactions (e.g., liver: de novo lipogenesis; muscle: fatty acid oxidation)
Metabolic state standardization:
Standardize feeding/fasting conditions before sample collection
Document glucose levels, insulin status, and other metabolic parameters
Consider time of day effects on metabolic signaling (circadian regulation)
Complementary techniques integration:
Combine antibody-based detection with functional assays (ACC activity, fatty acid synthesis rates)
Consider metabolomics approaches to measure pathway outputs (malonyl-CoA, fatty acids)
Use genetic models (AMPK knockout, ACC knock-in with S79A mutation) to validate pathway regulation
These considerations ensure that antibody-based detection of ACACA phosphorylation status accurately reflects the biological regulation of the AMPK-ACC pathway.
Preserving ACACA phosphorylation status during sample preparation requires specific protocols to inhibit phosphatase activity and maintain phosphoepitope integrity:
Immediate sample processing protocol:
Process tissues or cells immediately after collection
For tissues, flash-freeze in liquid nitrogen immediately after dissection
For cultured cells, avoid PBS washing steps that activate phosphatases; directly add lysis buffer to the plate
Optimized lysis buffer formulation:
Include multiple phosphatase inhibitors: sodium fluoride (50mM), sodium pyrophosphate (10mM), β-glycerophosphate (25mM), sodium orthovanadate (2mM)
Add protease inhibitor cocktail to prevent degradation
Use RIPA or modified RIPA buffer for better solubilization of membrane-associated proteins
Maintain cold temperature (4°C) throughout extraction
Physical disruption method selection:
For tissues: Pulverize frozen tissue under liquid nitrogen before adding to lysis buffer
For cells: Scrape cells directly into lysis buffer rather than trypsinizing
Avoid excessive sonication which can generate heat and activate phosphatases
Temperature control throughout processing:
Maintain samples on ice at all times during processing
Pre-chill all equipment (homogenizers, centrifuges)
Add ice-cold acetone for protein precipitation if concentrated samples are needed
Phosphatase inhibitor strategy:
Use fresh phosphatase inhibitors in all buffers
Consider adding okadaic acid (PP2A inhibitor) and calyculin A (PP1 inhibitor) for enhanced phosphatase inhibition
Include EDTA (1-2mM) to inhibit metal-dependent phosphatases
Sample storage considerations:
Add 2X Laemmli buffer and heat immediately after quantification for Western blot samples
For immunoprecipitation, maintain phosphatase inhibitors in all wash buffers
For long-term storage, add glycerol (final 10-20%) and store at -80°C
Validation of phosphorylation preservation:
Process aliquots of the same sample with and without phosphatase inhibitors as controls
Include positive controls (AMPK activator-treated samples) in each experiment
Monitor other phosphoproteins (e.g., phospho-AMPK) as internal controls for phosphatase inhibition efficacy
These optimized sample preparation protocols ensure that the phosphorylation status of ACACA at Ser79 accurately reflects the biological state at the time of sample collection.
When applying ACACA (Ab-79) antibodies to non-human samples, systematic assessment of cross-reactivity is essential for valid data interpretation:
Sequence homology analysis:
Epitope conservation evaluation:
Focus specifically on the phosphorylation site microenvironment
Even single amino acid differences can significantly affect antibody binding
Assess both primary sequence and potential structural conservation
Species validation strategy:
Graduated cross-reactivity testing:
Start with closely related species (primates, then other mammals)
Use increasing antibody concentrations to detect lower-affinity binding
Document species-specific optimal antibody dilutions
Multiple antibody comparison approach:
Test multiple antibodies targeting different epitopes of ACACA
Compare results between phospho-specific and total protein antibodies
Consider using species-specific secondary antibodies to reduce background
Positive and negative control selection:
Obtain species-specific positive controls (AMPK activator-treated samples)
Include samples from ACACA knockout models or siRNA-treated cells when available
Consider using recombinant proteins from target species as standards
Cross-validation with orthogonal methods:
Confirm antibody results with mass spectrometry-based phosphoproteomics
Use metabolic labeling to track ACC activity independent of antibody detection
Apply gene expression analysis to correlate protein detection with transcript levels
This methodical cross-reactivity assessment creates confidence in applying ACACA (Ab-79) antibodies beyond their validated human reactivity, potentially expanding their research utility while maintaining data reliability.
Designing experiments to investigate the relationship between ACACA phosphorylation and metabolic regulation requires a multi-faceted approach:
Stimulus-response experimental design:
Apply metabolic stimuli known to affect ACACA: insulin (decreases Ser79 phosphorylation), AMPK activators like AICAR or metformin (increase phosphorylation)
Design time-course experiments (5min, 15min, 30min, 1h, 3h, 24h) to capture both acute signaling and adaptive responses
Include dose-response studies to establish threshold effects
Cell/tissue model selection strategy:
Choose models based on metabolic relevance: hepatocytes (lipogenesis), adipocytes (fat storage), myocytes (fatty acid oxidation)
Consider primary cells vs. cell lines (primary cells offer more physiological responses)
For in vivo studies, target metabolically active tissues (liver, muscle, adipose) with appropriate controls
Pathway integration analysis:
Simultaneously monitor related pathways: insulin/AKT/mTOR (anabolic) and AMPK (catabolic)
Track multiple phosphorylation sites on ACACA (not just Ser79)
Measure upstream regulators (AMPK-Thr172 phosphorylation) and downstream metabolites (malonyl-CoA)
Genetic modification approach:
Use phospho-mutant models: ACACA-S79A (non-phosphorylatable) or S79D (phosphomimetic)
Apply CRISPR/Cas9 to generate endogenous mutations at the Ser79 site
Consider inducible systems to distinguish developmental from acute effects
Functional output measurement:
Directly measure ACACA enzymatic activity using radiometric or coupled assays
Track de novo lipogenesis using labeled substrates (¹⁴C-acetate incorporation)
Monitor fatty acid oxidation rates in parallel to assess metabolic switching
Nutrient manipulation design:
Apply glucose deprivation/refeeding protocols to modulate AMPK activity
Test high-fat vs. high-carbohydrate conditions to observe differential regulation
Include fasting/feeding cycles in in vivo studies to capture physiological regulation
Multi-omics integration:
Combine phosphoproteomics, metabolomics, and transcriptomics
Correlate ACACA phosphorylation with global metabolic profiles
Use metabolic flux analysis to determine the functional impact of phosphorylation changes
This comprehensive experimental design strategy enables researchers to establish causal relationships between ACACA phosphorylation status and metabolic outcomes, providing mechanistic insights into this critical regulatory node.
Optimizing Western blot conditions for ACACA (Ab-79) antibody detection requires attention to several technical parameters:
Sample preparation specifications:
Use RIPA buffer with phosphatase inhibitors for protein extraction
Load 20-50μg total protein per lane (ACACA is moderately abundant in metabolic tissues)
Include reducing agents (β-mercaptoethanol or DTT) in sample buffer
Heat samples at 70°C for 10 minutes rather than boiling to preserve phosphoepitopes
Gel selection and separation parameters:
Transfer optimization protocol:
For large proteins like ACACA (280kD), use wet transfer rather than semi-dry
Add 0.1% SDS to transfer buffer to improve high MW protein transfer
Extend transfer time (overnight at low voltage or 2-3 hours at higher voltage)
Use PVDF membrane (0.45μm pore size) rather than nitrocellulose for better protein retention
Blocking strategy selection:
For phospho-specific detection, use 5% BSA in TBST rather than milk (milk contains phosphatases)
For total ACACA detection, 5% non-fat dry milk in TBST is suitable
Block for 1-2 hours at room temperature or overnight at 4°C
Antibody dilution optimization:
Start with manufacturer's recommended dilution (typically 1:1000)
Perform a dilution series to determine optimal signal-to-noise ratio
Dilute antibodies in the same buffer used for blocking
Incubate primary antibody overnight at 4°C with gentle agitation
Washing protocol enhancement:
Perform 4-5 washes with TBST, 5-10 minutes each
Increase washing stringency (higher Tween-20 concentration, up to 0.1%) if background is high
Maintain consistent washing protocols between experiments
Detection system selection:
For phospho-specific detection, enhanced chemiluminescence (ECL) offers good sensitivity
Consider fluorescent secondary antibodies for multiplex detection and better quantification
Optimize exposure times to ensure signal is within linear range
Controls inclusion:
Run phosphatase-treated samples as negative controls
Include AMPK activator-treated samples as positive controls
Consider using recombinant ACACA protein as reference standard
Following these optimized conditions will maximize detection specificity and sensitivity when using ACACA (Ab-79) antibodies in Western blotting applications.
While immunoprecipitation (IP) is not listed among the validated applications for the ACACA (Ab-79) antibodies in the provided data , researchers can develop and optimize IP protocols with these considerations:
Antibody selection criteria:
Polyclonal antibodies targeting total ACACA may be more effective for IP than phospho-specific antibodies
If using phospho-specific antibodies, maintain phosphatase inhibitors throughout all steps
Consider using antibodies specifically validated for IP applications when available
Lysis buffer formulation:
Use mild non-denaturing buffers to preserve protein conformation and complexes
Standard IP buffer: 50mM Tris-HCl pH 7.5, 150mM NaCl, 1% NP-40, 0.5% sodium deoxycholate with protease inhibitors
For phospho-IP, add phosphatase inhibitors: 50mM NaF, 10mM Na₄P₂O₇, 2mM Na₃VO₄
Pre-clearing strategy:
Pre-clear lysates with appropriate control IgG and protein A/G beads
Perform for 1 hour at 4°C to reduce non-specific binding
Remove any precipitated material by centrifugation before adding the specific antibody
Antibody binding optimization:
Use 2-5μg antibody per 500μg-1mg total protein
Incubate overnight at 4°C with gentle rotation
Consider a crosslinking step to covalently attach antibody to beads (prevents antibody co-elution)
Bead selection and handling:
For rabbit antibodies, use protein A or protein A/G beads
Add pre-washed beads after antibody incubation
Use magnetic beads for gentler washing and better recovery
Washing stringency titration:
Perform sequential washes with increasing stringency
Start with lysis buffer, then add higher salt concentration (300mM NaCl)
Final washes with buffer without detergents
Keep all steps at 4°C to maintain complex integrity
Elution method selection:
For Western blot analysis: elute directly in 2X Laemmli buffer at 70°C
For activity assays: use peptide competition or mild elution conditions
For mass spectrometry: consider on-bead digestion to reduce contamination
Validation approaches:
Perform reciprocal IP with antibodies against known ACACA-interacting proteins
Include IgG control IP processed identically to experimental samples
Verify specificity by IP from ACACA-depleted or knockout samples when available
By adapting these methodological considerations, researchers can develop effective IP protocols using ACACA (Ab-79) antibodies for studying protein interactions and complexes involving ACACA.
ACACA (Ab-79) antibody can be powerful for investigating metabolic disease models when applied with these methodological considerations:
Disease model selection strategy:
Choose models relevant to ACACA function: insulin resistance, obesity, non-alcoholic fatty liver disease, cancer
Include appropriate controls matched for age, sex, and genetic background
Consider both genetic models (db/db, ob/ob mice) and diet-induced models (high-fat diet)
Tissue sampling prioritization:
Prioritize metabolically active tissues: liver (major site of de novo lipogenesis), adipose tissue, skeletal muscle
Consider tissue-specific differences in ACACA expression and regulation
Sample multiple adipose depots separately (subcutaneous, visceral, brown) as they have distinct metabolic profiles
Analytical approach development:
Measure both phospho-ACACA (Ser79) and total ACACA levels
Calculate phospho-to-total ratio to normalize for expression changes
Correlate with upstream regulators (phospho-AMPK) and downstream effects (fatty acid synthesis rates)
Context-specific interpretation framework:
In insulin resistance: expect decreased ACACA phosphorylation at baseline, with impaired response to AMPK activators
In obesity: analyze both basal phosphorylation and dynamic responses to feeding/fasting
In cancer models: assess metabolic reprogramming through altered ACACA regulation
Intervention response assessment:
Use ACACA phosphorylation as a biomarker for drug efficacy (e.g., metformin, AMPK activators)
Track acute vs. chronic changes in phosphorylation status
Correlate phosphorylation changes with physiological outcomes (glucose tolerance, insulin sensitivity)
Multi-parameter data integration:
Combine ACACA phosphorylation data with metabolic parameters (glucose, insulin, lipids)
Correlate with histological assessment of tissue lipid accumulation
Integrate with gene expression data for key lipogenic enzymes
Translational relevance establishment:
Compare findings between animal models and human samples when available
Consider species differences in metabolic regulation
Develop standardized protocols that can be applied across model systems
This methodological framework enables researchers to effectively use ACACA (Ab-79) antibody to gain mechanistic insights into metabolic disease pathophysiology and potential therapeutic interventions.
Quantifying ACACA phosphorylation in complex tissue samples requires specialized approaches to ensure accurate and reproducible results:
Sample preparation method selection:
Rapidly collect and process tissues to prevent post-collection changes in phosphorylation
Consider tissue-specific extraction protocols (e.g., liver requires different detergent composition than adipose)
Perform subcellular fractionation to enrich for cytosolic fraction where ACACA is predominantly located
Include phosphatase inhibitors at all stages of sample preparation
Normalization strategy optimization:
Implement a two-tier normalization approach:
First normalize phospho-ACACA to total ACACA
Then normalize to an appropriate loading control or total protein stain
Avoid exclusive reliance on single housekeeping proteins, which may vary in expression
Consider using total protein stains (SYPRO Ruby, Ponceau S) for robust normalization
Quantification method selection:
For Western blot analysis:
Use digital image capture systems with wide dynamic range
Ensure exposures fall within the linear range of detection
Apply consistent background subtraction methods
For ELISA-based detection:
Develop sandwich ELISA with capture antibody against total ACACA and detection antibody against phospho-Ser79
Include standard curves with recombinant phosphorylated and non-phosphorylated proteins
Statistical approach development:
Account for intra-tissue heterogeneity by analyzing multiple samples per tissue
Apply appropriate statistical tests based on data distribution
Consider power analysis to determine required sample size for detecting meaningful differences
Report both absolute and relative changes in phosphorylation
Alternative technology consideration:
Mass spectrometry-based phosphoproteomics for absolute quantification
Capillary Western systems (Wes, Jess) for improved reproducibility and lower sample requirements
Phospho-flow cytometry for cell-specific analysis in mixed cell populations
Proximity ligation assay for in situ detection of phosphorylated proteins
Validation controls inclusion:
Process paired samples with and without phosphatase inhibitors
Include calibrator samples on each gel/blot for inter-assay normalization
Run pooled reference samples across multiple experiments to assess reproducibility
Reporting standards implementation:
Document all sample processing steps with precise timing
Report antibody dilutions, exposure times, and image processing parameters
Present both representative images and quantitative data with appropriate statistical analysis
These methodological approaches enhance the accuracy and reliability of ACACA phosphorylation quantification in complex tissue samples, yielding more robust insights into metabolic regulation.
ACACA (Ab-79) antibody offers valuable tools for investigating the complex relationship between metabolism and cancer, with these methodological considerations:
Cancer model selection strategy:
Focus on cancers with known metabolic reprogramming (breast, prostate, colorectal, hepatocellular carcinoma)
Include both in vitro cancer cell lines and in vivo tumor models
Compare metabolic tissues (liver, adipose) with cancers arising from these tissues
Experimental design considerations:
Analyze ACACA phosphorylation across cancer progression stages
Compare cancer cells with normal cells of the same tissue origin
Assess changes in ACACA regulation under hypoxia, nutrient restriction, and other tumor microenvironment conditions
Mechanistic pathway analysis approach:
Investigate AMPK-ACC axis regulation in cancer contexts
Explore how oncogenic signaling (PI3K/AKT/mTOR) affects ACACA phosphorylation
Assess how altered ACACA regulation contributes to lipid synthesis required for cancer cell proliferation
Therapeutic intervention assessment:
Use ACACA phosphorylation as a pharmacodynamic marker for drugs targeting cancer metabolism
Test combination therapies targeting both ACACA and related metabolic enzymes
Monitor changes in ACACA phosphorylation in response to standard chemotherapies
Metabolic flux integration:
Correlate ACACA phosphorylation with de novo lipogenesis rates using isotope tracing
Assess how ACACA phosphorylation affects fatty acid oxidation in cancer cells
Monitor changes in lipid composition associated with altered ACACA activity
Single-cell level analysis:
Apply immunofluorescence with phospho-ACACA antibodies to assess heterogeneity within tumors
Combine with markers of proliferation, hypoxia, or stem-like properties
Develop protocols for phospho-flow cytometry to quantify ACACA phosphorylation at single-cell resolution
Translational research applications:
Evaluate ACACA phosphorylation in patient-derived xenografts or organoids
Assess potential as a biomarker for stratifying patients for metabolism-targeting therapies
Correlate ACACA phosphorylation patterns with patient outcomes and treatment responses
This research framework enables investigators to leverage ACACA (Ab-79) antibody for gaining insights into the role of altered metabolic regulation in cancer development, progression, and treatment response.
Implementing ACACA (Ab-79) antibody in multi-parameter immunofluorescence requires specific technical considerations:
Antibody compatibility assessment:
Test for cross-reactivity between antibodies from different host species
Ensure secondary antibodies do not cross-react with other primaries in the panel
Consider using directly conjugated primary antibodies to reduce species limitations
Signal amplification strategy selection:
For phospho-specific detection, implement tyramide signal amplification (TSA)
Consider quantum dots for narrow emission spectra and minimal overlap
Use zenon labeling technology for same-species antibody combinations
Protocol optimization approach:
Determine optimal fixation method (paraformaldehyde concentration, duration)
Optimize antigen retrieval conditions (heat-induced vs. enzymatic)
Test different permeabilization methods for accessing intracellular epitopes
Establish appropriate blocking conditions to minimize background
Sequential staining design:
Start with the weakest signal (often phospho-epitopes)
Implement sequential rather than simultaneous staining for challenging combinations
Consider antibody stripping and reprobing methods with appropriate controls
Controls implementation:
Include single-stained controls for each antibody to assess bleed-through
Use phosphatase-treated sections as negative controls for phospho-antibodies
Implement fluorescence-minus-one (FMO) controls to set thresholds
Image acquisition optimization:
Use spectral imaging to separate overlapping fluorophores
Apply consistent exposure settings across experimental groups
Establish threshold settings based on negative controls
Implement z-stack imaging for thick specimens
Quantification strategy development:
Develop automated image analysis workflows for unbiased quantification
Use nuclear counterstains for cell identification and normalization
Implement cell segmentation algorithms for single-cell analysis
Quantify both signal intensity and subcellular localization
Subcellular localization analysis:
Co-stain with markers for specific subcellular compartments
Assess ACACA distribution between cytosolic and membrane-associated pools
Examine co-localization with other metabolic enzymes or signaling molecules
These methodological considerations enable researchers to effectively incorporate ACACA (Ab-79) antibody into multi-parameter immunofluorescence studies, providing spatial context to ACACA regulation and function within cellular architecture.