ATX1 in Arabidopsis is a histone H3 lysine 4 trimethyltransferase (H3K4me3) regulating gene expression during development and stress responses .
Mechanism: ATX1 binds RNA Polymerase II (Pol II) and phosphatidylinositol 5-phosphate (PI5P), linking chromatin modification with lipid signaling .
Gene Targets: Modulates ~1,640 genes, including stress-responsive (WRKY70, LTP7) and developmental regulators .
Subcellular Localization: Shuttles between nucleus and cytoplasm, with PI5P binding enhancing nuclear retention .
ATOX1 is a copper chaperone delivering cytosolic copper to ATP7A/B transporters.
ATXN1 is linked to spinocerebellar ataxia type 1 (SCA1), caused by polyglutamine expansion.
ab186393 (Abcam): Mouse monoclonal, detects ~86 kDa ATXN1 in humans .
OAAJ02634 (Aviva): Rabbit polyclonal, phospho-specific (Ser775) .
#2177 (Cell Signaling): Detects endogenous ATXN1 at 105 kDa .
ATOX1 in Copper Homeostasis:
ATXN1 in Neurodegeneration:
Mechanism | Experimental Model | Outcome | Citations |
---|---|---|---|
Aggregation | Drosophila SCA1 model | Sequesters SMRTER, enhances toxicity | |
Transcriptional Repression | HEK-293 cells | Gal4-ATXN1 fusion represses promoters |
Neuroscience: Diagnosing SCA1 and analyzing ATXN1 aggregation .
Metallobiology: Tracking copper chaperone activity in disease models .
The following studies provide further evidence supporting the diverse roles of ATX1:
ATX1/ATXN1, or Ataxin-1, exists in two distinct research contexts. In human neurobiology, ATXN1 is a protein associated with spinocerebellar ataxia type 1, belonging to the polyglutamine (polyQ) expanded protein family . The pathological variant contains 41-81 CAG repeats compared to 6-39 in the normal allele . Mutations in this protein lead to progressive degeneration of the cerebellum, brain stem, and spinal cord.
In plant biology, ATX1 functions as a histone methyltransferase involved in secondary cell wall formation in Arabidopsis and as a copper distribution protein in Chlamydomonas . These distinct roles make ATX1 antibodies valuable tools in both neurological and plant research fields.
Based on current literature, researchers can access several types of ATX1 antibodies:
The choice of antibody depends on experimental requirements, including targeted post-translational modifications (like S776 phosphorylation) and the specific model organism under investigation.
Verifying antibody specificity is crucial for reliable research outcomes. A methodological approach includes:
Perform Western blot analysis of wild-type samples alongside known knockdown or knockout models (as demonstrated with the ATX1 amiRNA lines in Chlamydomonas ).
Include negative controls using empty vector-transformed cells or tissue samples.
Conduct immunoprecipitation followed by mass spectrometry to confirm target protein identity.
For immunofluorescence applications, compare staining patterns with GFP-tagged ATX1 expression to confirm localization patterns .
When working with phospho-specific antibodies (like ATXN1-S776), include dephosphorylated controls to validate signal specificity .
ATXN1 phosphorylation, particularly at serine 776 (S776), significantly influences protein function and disease pathogenesis. To investigate this aspect:
Employ phospho-specific antibodies that recognize ATXN1-S776 alongside total ATXN1 antibodies to quantify phosphorylation ratios in various experimental conditions.
Implement immunofluorescence using anti-ATXN1-S776 to visualize subcellular distribution of phosphorylated protein. The confocal immunofluorescence approach used in HeLa cells demonstrates phospho-ATXN1 localization when followed by Alexa Fluor 488-conjugated secondary antibodies .
Combine co-immunoprecipitation with phospho-specific antibodies to identify phosphorylation-dependent protein interactions, similar to the approach used to demonstrate ATXN1 interaction with endogenous U2AF65 .
Design experiments comparing wild-type ATXN1 with phospho-mimetic (S776D) and phospho-deficient (S776A) mutants to distinguish the functional impact of this modification on protein aggregation, localization, and interaction networks.
These approaches provide mechanistic insights into how phosphorylation modulates ATXN1 function in both normal and disease states.
Investigating the functional consequences of CAG repeat expansion in ATXN1 requires specialized approaches:
Comparative protein interaction studies: Implement immunoprecipitation with flag-tagged non-expanded (30Q) and expanded (82Q) ATXN1 constructs to identify differential binding partners, as demonstrated in the U2AF65 interaction studies .
Subcellular localization analysis: Utilize confocal microscopy with RFP-tagged ATXN1 constructs to compare localization patterns between normal and expanded variants. Prior research has shown both co-localize with endogenous U2AF65, despite differences in aggregation propensity .
Aggregation assays: Develop quantitative methods to measure aggregate formation kinetics using immunofluorescence with ATXN1 antibodies, comparing wild-type and expanded polyQ variants under various cellular stress conditions.
Proteostasis impact assessment: Combine ATXN1 antibodies with markers of cellular stress responses to evaluate how expanded ATXN1 affects global protein homeostasis networks.
These methodological approaches facilitate understanding of how polyQ expansion alters ATXN1 normal function and promotes neurodegeneration.
ATX1 in plants exhibits distinct nuclear and cytoplasmic activities, requiring specialized methodological approaches:
Subcellular fractionation: Separate nuclear and cytoplasmic fractions from plant cells followed by Western blotting with anti-ATX1 antibodies to quantify distribution between compartments .
Live-cell imaging: Utilize YFP-ATX1 fusion proteins combined with immunofluorescence using ATX1 antibodies to validate localization patterns in vivo, as implemented in Chlamydomonas studies .
Domain-specific antibodies: Generate antibodies targeting different domains (e.g., SET domain vs. N-terminal regions) to distinguish between full-length ATX1 (~22 kDa soloSET domain fragment) as observed in Arabidopsis .
Chromatin immunoprecipitation (ChIP): Apply ATX1 antibodies in ChIP experiments to identify genomic binding sites associated with its histone methyltransferase activity, particularly relevant to its role in regulating secondary cell wall formation .
These approaches help delineate the distinct functions of ATX1 in different cellular compartments and developmental contexts in plant systems.
Preserving antibody functionality requires careful attention to storage and handling protocols:
To maximize antibody performance, minimize freeze-thaw cycles by preparing single-use aliquots, avoid prolonged exposure to room temperature, and follow manufacturer-specific recommendations for reconstitution and dilution.
Robust experimental design requires appropriate controls:
For Western blotting:
Positive control: Cell/tissue lysate known to express ATX1/ATXN1 (e.g., CEM cell line for human ATXN1 )
Negative control: Lysate from ATX1 knockout or knockdown samples (e.g., atx1 mutant or amiRNA lines )
Loading control: Housekeeping protein detection to normalize expression levels
Specificity control: Pre-incubation of antibody with immunizing peptide to confirm signal specificity
Molecular weight verification: Confirmation that detected bands match expected sizes (e.g., full-length ATXN1 at 87 kDa and plant soloSET domain at ~22 kDa )
For immunofluorescence:
Secondary antibody-only control to assess background
Comparison with fluorescent protein-tagged constructs (GFP-ATX1) to validate localization patterns
DAPI nuclear counterstain to determine subcellular localization
Known expression pattern control (e.g., endogenous U2AF65 co-localization with ATXN1 )
These controls ensure experimental reliability and facilitate accurate interpretation of results.
Validating antibody performance in genetic models requires systematic approaches:
Expression confirmation in knockdown models: As demonstrated with ATX1 amiRNA lines in Chlamydomonas, confirm reduced protein levels correlate with decreased transcript abundance (50% and 25% reduction in respective lines) .
CRISPR knockout validation: In CRISPR/CPF1-edited ATX1 knockout lines, verify elimination of specific signal in Western blots, as shown in Chlamydomonas atx1-1 and atx1-2 models with introduced stop codons .
Domain-specific detection: When working with truncated variants or domain-specific functions, use antibodies targeting specific regions (N-terminal vs. SET domain) to differentiate between expression of full-length protein and functional fragments .
Cross-validation with heterologous expression: Express tagged versions (flag-tagged, GFP-fused) in cell culture systems to compare endogenous and exogenous protein detection by immunoblotting .
Tissue-specific expression analysis: Confirm antibody detects expected expression patterns in relevant tissues (e.g., interfascicular fiber cells for plant ATX1 involved in secondary cell wall formation) .
These validation steps ensure that experimental observations reflect genuine biological phenomena rather than technical artifacts.
When encountering detection challenges, consider these methodological solutions:
Signal enhancement:
Extend primary antibody incubation time (overnight at 4°C rather than 1 hour at room temperature)
Optimize blocking conditions (try 3% milk in PBS with 0.1% Tween 20 as used successfully for ATX1 detection )
Increase antibody concentration incrementally (while monitoring background)
Use signal amplification systems (e.g., avidin-biotin or tyramide signal amplification)
Background reduction:
Specificity improvement:
These adjustments help optimize signal-to-noise ratio while maintaining detection specificity.
Detecting low-abundance ATX1 variants requires specialized approaches:
Sample enrichment techniques:
Implement immunoprecipitation before Western blotting to concentrate target protein
Fractionate samples to isolate compartment-specific pools (nuclear vs. cytoplasmic)
Use phospho-enrichment techniques when studying phosphorylated ATXN1-S776
Detection system optimization:
Protocol adjustments:
Culture condition manipulation:
These approaches increase sensitivity while maintaining specificity for low-abundance ATX1 forms.
Recent methodological innovations expand the utility of ATX1 antibodies:
High-throughput screening applications: Develop immunoassay-based screens to identify compounds that modify ATXN1 phosphorylation, aggregation, or protein-protein interactions for therapeutic development.
Patient-derived model validation: Apply ATX1 antibodies to validate iPSC-derived neuronal models from spinocerebellar ataxia patients, comparing protein expression, phosphorylation, and localization with control cells.
Biomarker development: Investigate phosphorylated ATXN1-S776 levels in accessible patient samples as potential diagnostic or progression biomarkers.
Therapeutic target validation: Use ATX1 antibodies to monitor protein levels and modifications in response to experimental therapeutics, including antisense oligonucleotides or small molecule interventions.
Evolutionary conservation studies: Apply antibodies across species to investigate functional conservation of ATX1/ATXN1 between plants and animals, potentially revealing fundamental biological principles.
These emerging applications extend beyond traditional descriptive studies to actively contribute to translational research and therapeutic development.
Integrating ATX1 antibodies into multi-omics frameworks offers powerful research opportunities:
Proximity labeling applications: Couple ATX1 antibodies with BioID or APEX2 systems to identify proximal proteins in living cells, expanding our understanding of the dynamic ATX1 interactome.
Spatial transcriptomics integration: Combine immunofluorescence using ATX1 antibodies with in situ sequencing to correlate protein localization with local transcriptional landscapes.
Single-cell proteomics: Implement ATX1 antibodies in mass cytometry (CyTOF) or single-cell Western blotting to investigate cellular heterogeneity in ATX1 expression and modification states.
Structural biology applications: Use antibody fragments (Fabs) as crystallization chaperones to facilitate structural studies of difficult-to-crystallize ATX1 domains or complexes.
In vivo imaging development: Develop site-specifically labeled ATX1 antibodies for in vivo imaging of protein dynamics in model organisms.
These innovative approaches extend antibody applications beyond conventional biochemical and histological techniques, enabling comprehensive multi-dimensional analysis of ATX1 biology.