Research highlights ACO2's involvement in multiple pathological conditions:
ACO2 deficiency exacerbates Parkinson’s disease (PD) pathology by promoting mitochondrial dysfunction and dopaminergic neuron degeneration. Knockdown models show reduced autophagy-related gene expression (e.g., LC3, Atg5) and impaired energy metabolism .
Decreased ACO2 activity is observed in Huntington’s disease (HD) patients, linking mitochondrial dysfunction to neurodegeneration .
Pan-Cancer Analysis: ACO2 is overexpressed in 16 tumor types (e.g., liver, breast, lung cancers) and downregulated in 5 (e.g., glioblastoma, kidney cancer). Its expression correlates with immune infiltration (e.g., CD8+ T cells, tumor-associated neutrophils) and prognosis .
Functional Role: ACO2 promotes hepatocellular carcinoma (HCC) proliferation and migration. Lipidomics reveals altered metabolite profiles (e.g., reduced phosphatidylcholine levels) upon ACO2 knockdown .
ACO2 suppresses antibacterial immunity by modulating oxaloacetate levels and mitochondrial unfolded protein response (UPRmt). Inhibition of ACO2 enhances survival in C. elegans and human cells infected with pathogens like S. aureus .
Key validation data from commercial antibodies:
ACO2 knockdown in C. elegans and HeLa cells increases susceptibility to bacterial pathogens by disrupting UPRmt and oxaloacetate metabolism .
ACO2 inhibition enhances immune cytokine production (e.g., IL-6, IL-8) in human cells, suggesting potential for immunomodulatory therapies .
ACO2’s diagnostic value is highlighted in six tumor types, with correlations to immune checkpoint genes (e.g., PD-L1) and tumor mutational burden (TMB) .
ACO2 antibodies are available with various specifications to meet different research needs. Most commonly, these antibodies target different regions of the ACO2 protein and come in different formats based on host and clonality. For example, the ABIN357262 antibody targets the central region of human ACO2, is raised in rabbits as a polyclonal antibody, and demonstrates reactivity with human, rat, and mouse samples . This antibody is suitable for Western Blotting (WB) and Enzyme Immunoassay (EIA) applications.
The table below summarizes the common ACO2 antibody variants available for research:
When selecting an ACO2 antibody for your research, consider these key factors:
Experimental application: Different ACO2 antibodies are optimized for specific techniques. For protein detection and quantification, select antibodies validated for Western blotting. For localization studies, choose antibodies validated for immunohistochemistry or immunofluorescence .
Species compatibility: Ensure the antibody has been validated in your species of interest. The search results indicate that many ACO2 antibodies work with human, rat, and mouse samples, but cross-reactivity varies by antibody .
Target region specificity: Select antibodies that target relevant regions of the ACO2 protein. For studies involving variants like A252T, ensure your antibody can detect the region containing or affected by this variant .
Clonality considerations: Polyclonal antibodies offer broader epitope recognition but may have batch-to-batch variability. Monoclonal antibodies provide higher specificity and consistency but may be less sensitive .
Application-specific validation: Review the validation data provided by manufacturers to ensure the antibody has been thoroughly tested for your specific application.
For optimal Western blotting results with ACO2 antibodies, follow these methodological guidelines:
Sample preparation:
For whole cell lysates, use RIPA buffer with protease inhibitors
For mitochondrial enrichment, consider differential centrifugation protocols
Maintain cold conditions throughout to preserve protein integrity
Electrophoresis parameters:
Use 10-12% SDS-PAGE gels appropriate for ACO2's molecular weight (~85 kDa)
Load 20-50 μg of total protein per lane
Include molecular weight markers spanning 50-100 kDa range
Transfer conditions:
Use PVDF membranes for optimal protein binding
Transfer at 100V for 60-90 minutes or 30V overnight at 4°C
Verify transfer efficiency with reversible protein staining
Antibody incubation:
Block membranes with 5% non-fat milk or BSA in TBS-T for 1 hour
Dilute primary ACO2 antibody according to manufacturer's recommendation (typically 1:500-1:2000)
Incubate with primary antibody overnight at 4°C
Use appropriate HRP-conjugated secondary antibody (1:2000-1:5000)
Controls to include:
Positive control (tissue with known ACO2 expression, such as liver)
Loading control (mitochondrial marker for mitochondrial fractions)
ACO2 knockdown samples as negative controls
ACO2 activity measurement is crucial for understanding its functional role in disease states. Research has shown that ACO2 activity is significantly decreased in peripheral blood mononuclear cells (PBMCs) from Parkinson's disease patients and correlates with disease progression . Here are methodological approaches:
Spectrophotometric assays:
Measure the conversion rate of citrate to isocitrate through cis-aconitate
Monitor absorbance changes at 240nm
Include proper controls and blanks to account for non-enzymatic reactions
Oxygen consumption measurement:
Sample considerations:
Data interpretation:
Research has revealed several crucial mechanisms connecting ACO2 dysfunction to Parkinson's disease pathogenesis:
Mitochondrial metabolism impairment:
Autophagy dysregulation:
α-synuclein interaction:
Genetic susceptibility:
These mechanisms collectively contribute to increased oxidative stress, energy deficiency, and neuronal vulnerability characteristic of Parkinson's disease.
The ACO2 A252T variant, identified in Parkinson's disease patients, demonstrates several functional alterations at cellular and molecular levels:
Enzymatic activity:
Mitochondrial respiration:
Mitochondrial membrane potential:
Autophagy function:
Epigenetic regulation:
Disease model effects:
These findings establish ACO2 A252T as a risk factor that increases vulnerability to Parkinson's disease, particularly when combined with environmental stressors.
To investigate the critical interaction between α-synuclein and ACO2, researchers can employ several experimental strategies:
In vivo models:
Co-localization studies:
Immunofluorescence microscopy using antibodies against ACO2 and α-synuclein
Super-resolution microscopy for detailed interaction analysis
Proximity ligation assays to confirm direct interaction in tissue samples
Protein interaction analysis:
Co-immunoprecipitation with ACO2 antibodies to pull down α-synuclein or vice versa
FRET (Förster Resonance Energy Transfer) to assess direct protein interactions in living cells
Surface plasmon resonance to measure binding kinetics between purified proteins
Functional consequence assessment:
Measure ACO2 activity in the presence of recombinant α-synuclein
Assess mitochondrial function parameters with combined ACO2/α-synuclein manipulation
Metabolite profiling to identify TCA cycle alterations resulting from the interaction
Research has demonstrated that targeting the α-synuclein-ACO2 interaction may represent a promising therapeutic strategy for improving mitochondrial function in Parkinson's disease .
ACO2 antibodies serve as powerful tools for investigating mitochondrial dysfunction across multiple experimental platforms:
Protein expression profiling:
Tissue and cellular localization:
Complex experimental paradigms:
Correlation studies:
Therapeutic evaluation:
Recent research has identified several promising therapeutic strategies targeting ACO2 for neurodegenerative disorders:
Gene therapy approaches:
Mitochondrial function enhancement:
Autophagy modulation:
Since ACO2 deficiency impairs autophagy via histone acetylation mechanisms, targeting these epigenetic pathways represents a potential approach
Histone deacetylase inhibitors might counteract the reduced H3K9 and H4K5 acetylation caused by ACO2 dysfunction
Autophagy activators could potentially bypass the ACO2-related defects
Biomarker applications:
These emerging strategies highlight ACO2 as a promising therapeutic target for improving mitochondrial function in Parkinson's disease and potentially other neurodegenerative disorders.
Understanding the relationship between ACO2 protein expression and enzymatic activity is crucial for accurate data interpretation:
Mechanistic explanations for discrepancies:
Interpretation framework:
Decreased activity with normal expression: Suggests functional inactivation, possibly due to oxidative damage or α-synuclein binding
Decreased expression and activity: Indicates transcriptional/translational downregulation
Increased expression with normal/low activity: Suggests compensatory upregulation in response to functional deficits
Methodological considerations:
Disease-specific patterns:
Research implications:
Proper controls and normalization strategies are critical for generating reliable and reproducible results with ACO2 antibodies:
Essential experimental controls:
Positive tissue controls: Liver, heart, or brain tissues with known ACO2 expression
Negative controls: ACO2 knockdown or knockout samples where available
Antibody controls: Secondary antibody-only controls to detect non-specific binding
Loading controls: Mitochondrial markers (e.g., VDAC, COX IV) for mitochondrial preparations
Disease model-specific controls:
Normalization strategies:
Validation approaches:
Statistical considerations:
These strategies help ensure that changes observed in ACO2 expression or function are genuine and disease-relevant rather than artifacts of experimental variation.