Phospho-ATXN1 (Ser776) Antibody

Shipped with Ice Packs
In Stock

Description

Biological Relevance of ATXN1-S776 Phosphorylation

Phosphorylation at S776 stabilizes ATXN1, influencing its pathogenicity in SCA1. Key findings include:

  • Stabilization Mechanism: Phosphorylated ATXN1 (pS776-ATXN1) exhibits reduced degradation compared to non-phosphorylated forms, contributing to toxic accumulation in cerebellar Purkinje cells .

  • Kinase Association: Cyclic AMP-dependent protein kinase (PKA), not Akt, is the primary kinase responsible for S776 phosphorylation in the cerebellum (Fig. 7, ).

  • Disease Modulation: Genetic or pharmacological inhibition of PKA reduces ATXN1-S776 phosphorylation, enhances ATXN1 degradation, and improves motor deficits in SCA1 mouse models .

Mechanistic Studies in SCA1

  • In vitro Kinase Assays: Cerebellar lysate-based assays confirmed PKA-mediated phosphorylation of GST-ATXN1-S776, with inhibition by staurosporine and PKA-specific peptides (Figs. 6–8, ).

  • Therapeutic Targeting: GSK690693 (a PKA inhibitor) reduced ATXN1 levels by 44% in SCA1 patient-derived neurons and 70% in mouse cerebellar slices after 5 days (Fig. 2B–C, ).

Diagnostic and Functional Insights

  • Subcellular Localization: pS776-ATXN1 is enriched in the nucleus, while its kinase activity resides in the cytoplasm, suggesting phosphorylation precedes nuclear translocation (Fig. 6D, ).

  • Post-Translational Crosstalk: S776 phosphorylation enhances ATXN1’s sumoylation and ubiquitination resistance, exacerbating toxicity in polyglutamine-expanded mutants .

Pharmacological Inhibition

CompoundEffect on ATXN1-S776Cellular SystemOutcome
StaurosporineReduced phosphorylation (IC₅₀: ~10 nM)DAOY cells, iPSC-derived neuronsRapid ATXN1 degradation (≤6 minutes)
GSK690693Decreased pS776 levelsMouse cerebellar slices70% reduction in ATXN1 after 5 days

Genetic Modulation

  • PKA Catalytic Subunit Cα Knockdown: In Pcp2-ATXN1[82Q] mice, reduced PKA activity lowered ATXN1[82Q]-S776 phosphorylation and delayed motor dysfunction .

Clinical and Research Implications

  • Biomarker Potential: pS776-ATXN1 levels correlate with disease progression in SCA1 models, offering a quantifiable readout for therapeutic screens .

  • Therapeutic Strategy: Targeting PKA-mediated phosphorylation could promote ATXN1 clearance, as evidenced by improved cerebellar function in preclinical studies .

Limitations and Considerations

  • Species Specificity: While reactive in human and mouse models, cross-reactivity in non-mammalian systems requires further validation .

  • Kinase Complexity: Despite PKA’s dominant role, contributions from other kinases in non-cerebellar tissues remain unexplored .

Product Specs

Form
Supplied at 1.0mg/mL in phosphate buffered saline (without Mg2+ and Ca2+), pH 7.4, 150mM NaCl, 0.02% sodium azide and 50% glycerol.
Lead Time
Typically, we can ship your orders within 1-3 business days of receipt. Delivery timelines may vary depending on the purchasing method and location. For specific delivery estimates, please consult your local distributors.
Synonyms
alternative ataxin1 antibody; Ataxin-1 antibody; ATX1 antibody; ATX1_HUMAN antibody; Atxn1 antibody; D6S504E antibody; OTTHUMP00000016065 antibody; SCA1 antibody; Spinocerebellar ataxia type 1 protein antibody
Target Names
Uniprot No.

Target Background

Function
Ataxin-1, a chromatin-binding factor, acts as a corepressor for CBF1, repressing Notch signaling in the absence of the Notch intracellular domain. It binds to the HEY promoter, potentially aiding in RBPJ-mediated repression alongside NCOR2. Ataxin-1 demonstrates RNA binding in vitro and may play a role in RNA metabolism. In conjunction with CIC and ATXN1L, it participates in brain development.
Gene References Into Functions
  1. Converging evidence identifies ATXN1 as a promising risk gene for Schizophrenia. The integrated approach employed here proves beneficial in dissecting the genetic basis of schizophrenia. PMID: 29055568
  2. JNK and DUSP18 exhibit reciprocal modulation of SUMOylation, which plays a regulatory role in ataxin-1 aggregation. PMID: 29852174
  3. ATXN1 might contribute to neuronal degeneration associated with ALS. PMID: 29274668
  4. SCA1 mutation carriers exhibited performance similar to controls in postural tasks with open eyes. However, in conditions lacking visual feedback, SCA1 carriers demonstrated significantly higher stability indices than controls across all longitudinal evaluations. Carriers closer to disease onset (PMID: 28551466
  5. In cervical cancer cells, ATXN1 knockdown induced EMT by directly regulating Snail expression, leading to matrix metalloproteinase activation and promotion of cell migration and invasion. PMID: 28212558
  6. ATXN1 underexpression has been linked to metabolic diseases. PMID: 27918534
  7. Data suggest that GSK3b and mTOR pathways modulate ATXN1 function in spinocerebellar ataxia type-1 (SCA1) pathogenesis. These pathways present potential therapeutic targets prior to the onset of disease symptoms in SCA1 and other pathologies involving ATXN1 dysregulation. PMID: 27466200
  8. SCA1 phenotypes were reversed by partial suppression of human mutant ATXN1 mRNA through rAAV.miS1 delivery after symptom onset in mice. PMID: 27686464
  9. SCA1 relative frequency in Poland is the highest compared to other countries worldwide in patients with Spinocerebellar ataxias. PMID: 27193757
  10. Molecular modeling was used to investigate protein-protein interactions contributing to the AXH domain dimer stability in Ataxin-1. PMID: 26879337
  11. Data indicate that in spinocerebellar ataxia type 1 patients, the spinocerebellar ataxia type 1 protein trinucleotide repeat expansion (CAG)n was greater than 39, compared to the normal range of 6-38. PMID: 27577232
  12. This study reports the results of molecular dynamics simulations of the AXH monomer of Ataxin-1. PMID: 26522012
  13. Systematic replacement of each lysine residue in the AXH domain revealed that the lysine at position 589 (K589) of ATXN1 is essential for its ubiquitylation by UbcH6. PMID: 25641559
  14. Results indicate that two SNPs in the ATXN1 gene exhibit a founder effect of the same repeat carrying allele as in the general Indian population, suggesting that Spinocerebellar ataxia type 1 disease onset is significantly delayed when transmission is maternal. PMID: 25344417
  15. This work provides the structural and molecular basis for the interaction between RBM17 and the phosphorylated form of ATXN1. PMID: 24858692
  16. Partner recognition of the AXH domain of the transcriptional co-regulator ataxin-1 is fine-tuned by a subtle balance between self- and hetero-associations. PMID: 24155902
  17. Cerebellar neurochemical alterations were measured in a knock-in mouse model of spinocerebellar ataxia type 1, a hereditary movement disorder, using ultra-high field magnetic resonance spectroscopy (MRS). PMID: 24032423
  18. Data indicate that the alternative ataxin-1 (ATXN1) protein is constitutively co-expressed and interacts with ATXN1. PMID: 23760502
  19. SCA 1 was the most prevalent type of SCA identified in the Autosomal dominant hereditary ataxia in Sri Lanka. PMID: 23634774
  20. Results demonstrate that variation in ATXN1 is implicated in disordered gambling. PMID: 22780124
  21. The AXH domain of ataxin-1 exhibits conformational heterogeneity. PMID: 23528090
  22. Patients with JARID2 deletion manifested with cognitive impairment, gait disturbance, and a characteristic facial appearance, whereas those with ATXN1 deletion displayed intellectual disability and behavioral abnormalities. PMID: 23294540
  23. Multiple components of the RAS-MAPK-MSK1 pathway influence ATXN1 levels in Drosophila and human cells. PMID: 23719381
  24. This study showed that progressive cerebellar degeneration requires expressing ATXN1 with an expanded polyglutamine tract. PMID: 23536093
  25. Both ATXN-1 and ATXN-2 PolyQ intermediate expansions are independently associated with an increased risk for ALS. PMID: 23197749
  26. MED15 and PUM1 proteins, possessing coiled-coil domains, act as potent enhancers of polyQ-mediated ataxin-1 protein misfolding and proteotoxicity in vitro and in vivo. PMID: 22916034
  27. FOX-2 is involved in splicing of ataxin-2 transcripts, and this splicing event is altered by ataxin-1 overexpression. PMID: 22666429
  28. Data indicate that neuroprotectin D1 (NPD1) synthesis is an early response induced by proteotoxic stress due to abnormally folded ataxin-1. PMID: 22511762
  29. Results suggest that expanded ATXN1 may induce unregulated ionic pathways in the nuclear membrane, causing significant cellular damage. PMID: 22330095
  30. This study demonstrated that the progression of autosomal dominant cerebellar ataxia and spastic paraplegia was faster in SCA s with polyglutamine expansions in SCA1, 2, and 3. PMID: 22491195
  31. Both ATXN1 and BOAT1 bind to the promoter region of Hey1 and inhibit Notch transcriptional output through direct interactions with CBF1, a crucial transcription factor for the Notch pathway. PMID: 21475249
  32. This study demonstrates that ataxin-1 occupies the promoter region of E-cadherin in vivo and activates the promoter in a CtBP2-mediated transcriptional regulation manner. PMID: 21315774
  33. The ATXN1 gene is associated with intelligence in individuals with an ADHD background. PMID: 21302343
  34. Phosphorylation of transgenic ataxin-1 at the serine-776 motif is crucial for ataxin-1-mediated toxicity. PMID: 20477910
  35. ATXN1 functions as a genetic risk modifier contributing to AD pathogenesis through a loss-of-function mechanism by regulating beta-secretase cleavage of APP and Abeta levels. PMID: 20097758
  36. The neurochemical alterations observed in SCA1[82Q] transgenic mice are primarily attributed to the expansion of the polyglutamine repeat in ataxin-1, rather than the overexpression of the human protein. PMID: 20220018
  37. Collectively, these findings indicate that SUMO modification of ataxin-1 promotes its aggregation and that oxidative stress and the JNK pathway play roles in this process. PMID: 20132795
  38. p62 contributed to the assembly of proteasome-containing degradative compartments in the vicinity of nuclear aggregates containing polyglutamine-expanded Ataxin1Q84 and to the degradation of Ataxin1Q84. PMID: 20018885
  39. In spinocerebellar ataxia, a trinucleotide expansion disease, the SCA1 allele with 31 repeats plays a potential role in triplet diseases. The SCA1 region may also be involved in pathological trinucleotide repeat expansions. PMID: 11807410
  40. Spinocerebellar ataxia type 1 (SCA1): phenotype-genotype correlation studies in intermediate alleles. PMID: 11973625
  41. Two types of ataxin1 inclusions exist: those undergoing rapid, complete exchange with a nucleoplasmic pool and those containing varying levels of slow-exchanging ataxin1. The latter inclusions display high ubiquitin levels but low proteasome levels. PMID: 12360291
  42. Phylogenetic evidence from deletions in SCA1. PMID: 12411613
  43. The p80 coilin protein co-localizes with ataxin-1 aggregates in the nucleoplasm. PMID: 12757932
  44. Analysis of the SCA1 AXH domain structure. PMID: 14583607
  45. Abolishing full-length mutant human ataxin-7 transgene expression did not reverse retinopathy progression in SCA7 mice, suggesting that some polyQ-induced pathological events may be irreversible. PMID: 14985428
  46. The structural role of the CAU interruptions in the SCA1 transcripts that destabilize the CAG repeat hairpin. PMID: 15292212
  47. SCA1 genotypes in a Polish population demonstrate significant differences in allele spectra and frequencies compared to other populations. The dynamic mutation of SCA1 may originate from the expansion of long pure repeat tracts without prior loss of interruptions. PMID: 15300851
  48. Ataxin-1 localization to inclusions and inclusion dynamics within the nucleus are RNA- and transcription-dependent. PMID: 15615787
  49. Polyglutamine-expanded ataxin-1 decreases proteasome activity, implying that a disruption in the ubiquitin-proteasome pathway is directly involved in the development of spinocerebellar ataxia type 1. PMID: 15750336
  50. A novel phosphorylation site at serine 239 was demonstrated in ataxin-1. PMID: 15878393

Show More

Hide All

Database Links

HGNC: 10548

OMIM: 164400

KEGG: hsa:6310

STRING: 9606.ENSP00000244769

UniGene: Hs.434961

Involvement In Disease
Spinocerebellar ataxia 1 (SCA1)
Protein Families
ATXN1 family
Subcellular Location
Cytoplasm. Nucleus.
Tissue Specificity
Widely expressed throughout the body.

Q&A

What is ATXN1 and why is phosphorylation at Ser776 significant?

ATXN1 is a chromatin-binding factor that functions as a repressor of Notch signaling and plays important roles in brain development. It is primarily known for its association with SCA1, an inherited neurodegenerative disorder caused by the expansion of a polyglutamine tract in the ATXN1 protein .

Phosphorylation of ATXN1 at serine 776 (Ser776) plays a crucial role in SCA1 pathogenesis. Research has demonstrated that this post-translational modification significantly affects protein stability, with phosphorylated ATXN1 being less susceptible to degradation . Studies in cerebellar Purkinje cells, a prominent site of SCA1 pathology, have confirmed that phosphorylation at Ser776 stabilizes the protein . Furthermore, this phosphorylation affects the pathogenicity of proteins with expanded polyglutamine tracts, directly influencing disease progression mechanisms .

What are the key characteristics of Phospho-ATXN1 (Ser776) antibodies?

Phospho-ATXN1 (Ser776) antibodies are specifically designed to detect ATXN1 protein only when phosphorylated at the Ser776 residue . These antibodies are available in multiple formats:

  • Rabbit polyclonal antibodies targeting regions around the phosphorylation site (typically amino acids 742-791)

  • Mouse monoclonal antibodies with similar specificity

Key specifications include:

  • Applications: Western blot (1:500-1:2000 dilution), immunohistochemistry (1:100-1:300), immunofluorescence (1:200-1:1000), and ELISA (1:10000)

  • Reactivity: Typically reactive with human and mouse ATXN1

  • Formulation: Usually supplied in PBS containing 50% glycerol, 0.5% BSA, and 0.02% sodium azide

  • Storage: Recommended at -20°C with avoidance of repeated freeze-thaw cycles

Importantly, these antibodies are designed to detect endogenous levels of ATXN1 protein only when phosphorylated at Ser776, making them valuable tools for studying this specific post-translational modification .

How can researchers validate the specificity of Phospho-ATXN1 (Ser776) antibodies?

Validating antibody specificity is crucial for generating reliable research data. For Phospho-ATXN1 (Ser776) antibodies, several approaches are recommended:

  • Peptide competition assays: Blocking with the phospho-peptide used as the immunogen should eliminate specific antibody binding. Western blot and immunofluorescence data show that signal disappears when the antibody is pre-incubated with the phosphopeptide, confirming phospho-specificity .

  • Phospho-resistant mutants: Using ATXN1 constructs with serine-to-alanine mutations at position 776 (S776A) provides an excellent negative control. These constructs cannot be phosphorylated at this position and should not be detected by phospho-specific antibodies .

  • Multiple detection methods: Validating antibody specificity across different applications (Western blot, immunofluorescence, immunohistochemistry) strengthens confidence in antibody performance .

  • Treatment conditions: Using samples with known phosphorylation status, such as HepG2 cells treated with Adriamycin (0.5μM for 5 hours), which has been shown to affect ATXN1 phosphorylation, provides positive controls .

These validation steps ensure that the observed signals truly represent phosphorylated ATXN1 rather than non-specific interactions or detection of non-phosphorylated forms.

What are the recommended experimental conditions for using these antibodies?

Optimal experimental conditions vary by application but generally include:

  • Western blot:

    • Protein loading: 20-50 μg of total protein per lane

    • Dilution range: 1:500-1:2000

    • Blocking: 5% BSA in TBST (BSA is often preferred over milk for phospho-epitopes)

    • Incubation: Overnight at 4°C for primary antibody

  • Immunohistochemistry:

    • Dilution range: 1:100-1:300

    • Antigen retrieval: Typically required for formalin-fixed tissues

    • Detection system: Biotin-streptavidin or polymer-based systems

  • Immunofluorescence:

    • Dilution range: 1:200-1:1000

    • Fixation: 4% paraformaldehyde recommended

    • Permeabilization: 0.1-0.5% Triton X-100

  • Critical factors across applications:

    • Sample preparation: Include phosphatase inhibitors in all extraction buffers

    • Controls: Include both positive controls (known phosphorylated samples) and negative controls

    • Storage: Maintain antibodies at -20°C and avoid repeated freeze-thaw cycles

Following these recommendations will help maximize signal specificity while minimizing background and non-specific binding.

Which kinase phosphorylates ATXN1 at Ser776 in cerebellar Purkinje cells?

The identity of the kinase responsible for ATXN1 phosphorylation at Ser776 has been a subject of investigation with evolving understanding. While earlier studies using transfected cell lines and Drosophila models suggested Akt (protein kinase B) as the responsible kinase, more recent evidence from cerebellar Purkinje cells points to cyclic AMP-dependent protein kinase (PKA) as the primary kinase .

Evidence supporting PKA as the ATXN1-S776 kinase includes:

  • Immunodepletion studies: Depletion of Akt from cerebellar extracts did not significantly affect phosphorylation of GST-ATXN1 at S776, whereas PKA immunodepletion substantially reduced this phosphorylation .

  • Fractionation experiments: PKA co-fractionated with ATXN1-S776 kinase activity in cerebellar cytosol under both ammonium sulfate precipitation (50-90% fraction) and hydroxyapatite chromatography conditions .

  • Inhibitor studies: PKA inhibitors (staurosporine and a 17-residue PKA inhibitor peptide) significantly reduced S776 phosphorylation in a dose-dependent manner, while an Akt inhibitor had no significant effect .

  • Subcellular localization: The kinase activity was found to be enriched in the cytoplasmic fraction, while phosphorylated ATXN1 accumulated in the nuclear fraction .

These findings highlight the importance of studying kinase-substrate relationships in physiologically relevant contexts, as results may differ between model systems.

How can researchers establish an in vitro ATXN1 phosphorylation assay?

An in vitro phosphorylation assay for ATXN1 can be established following these methodological steps:

  • Substrate preparation:

    • Generate recombinant GST-ATXN1 fusion proteins, including both wild-type (S776) and phospho-resistant mutant (A776) versions

    • The A776 mutant serves as an excellent negative control to confirm phosphorylation specificity

  • Kinase source preparation:

    • Prepare cerebellar extracts as a source of native kinase activity

    • For subcellular fractionation, the cytoplasmic fraction contains higher kinase activity than the nuclear fraction

    • Further fractionation can be performed using ammonium sulfate precipitation (50-90% fraction) followed by hydroxyapatite column chromatography with a sodium phosphate gradient

  • Phosphorylation reaction:

    • Incubate GST-ATXN1 with cerebellar extract under appropriate buffer conditions

    • The phosphorylation signal increases with increasing amounts of cerebellar extract, confirming enzyme-dependent phosphorylation

    • Include appropriate cofactors and ATP in the reaction buffer

  • Detection methods:

    • Western blotting with phospho-Ser776-specific antibodies is the primary detection method

    • The signal should increase with wild-type ATXN1 but remain minimal with the A776 mutant

  • Validation approaches:

    • Immunodepletion: Remove specific kinases (e.g., PKA) from the extracts to confirm their role

    • Inhibitor studies: Add kinase inhibitors to test their effects on phosphorylation

    • Peptide competition: Use phospho-peptides to validate antibody specificity

This assay system provides a powerful tool for mechanistic studies of ATXN1 phosphorylation and potential therapeutic interventions.

How does phosphorylation at Ser776 affect ATXN1 protein stability and function?

Phosphorylation of ATXN1 at Ser776 has profound effects on protein properties:

  • Protein stability: Research has demonstrated that phosphorylation at S776 stabilizes ATXN1 protein. Studies using phospho-resistant alanine mutations at residue 776 (S776A) show that these mutants are destabilized in Purkinje cells compared to wild-type (S776) ATXN1 . This stabilization effect may contribute to pathology by increasing the cellular burden of mutant ATXN1 protein.

  • Subcellular localization: While the kinase activity responsible for S776 phosphorylation is enriched in the cytoplasmic fraction, phosphorylated ATXN1 preferentially accumulates in the nuclear fraction . This suggests that phosphorylation may affect nucleocytoplasmic trafficking or nuclear retention of ATXN1.

  • Interaction with other post-translational modifications: Phosphorylation at S776 influences other modifications of ATXN1. For example:

    • Sumoylation of ATXN1 depends on both nuclear localization and phosphorylation at S776

    • Ubiquitination and subsequent proteasomal degradation of ATXN1, mediated by UBE3A, appears to be affected by phosphorylation status

  • Polyglutamine expansion effects: In SCA1 patients with expanded polyglutamine repeats in ATXN1, phosphorylation at S776 increases the pathogenicity of these expanded proteins . Additionally, the presence of expanded polyglutamine repeats impairs ubiquitination and degradation, leading to accumulation of ATXN1 in neurons and subsequent toxicity .

These findings highlight the multifaceted role of S776 phosphorylation in determining ATXN1 protein fate and function, particularly in the context of SCA1 pathogenesis.

What experimental challenges exist when studying ATXN1 phosphorylation?

Researchers face several technical and biological challenges when investigating ATXN1 phosphorylation:

  • Kinase identification discrepancies: Different experimental systems have yielded contradictory results regarding the responsible kinase. While Akt was implicated in cell lines and Drosophila models, PKA appears to be the primary kinase in cerebellar Purkinje cells . This highlights the importance of validating findings across different model systems.

  • Compartmentalization challenges: The finding that kinase activity is enriched in cytoplasmic fractions while phosphorylated ATXN1 accumulates in nuclear fractions complicates the study of the phosphorylation process . This spatial separation makes real-time visualization of the phosphorylation event challenging.

  • Technical considerations:

    • Preserving phosphorylation during sample processing requires careful use of phosphatase inhibitors

    • Antibody specificity requires thorough validation through multiple approaches

    • Quantification of phosphorylation stoichiometry remains technically demanding

  • Polyglutamine length variations: Different polyglutamine repeat lengths in ATXN1 (e.g., ATXN1[30Q] vs. ATXN1[82Q]) may exhibit different phosphorylation patterns or responses to manipulation , making cross-study comparisons complex.

  • Model system relevance: Translation between in vitro systems, cell cultures, and in vivo models requires careful consideration of the biological context and limitations of each approach.

Understanding these challenges is essential for designing robust experimental approaches and correctly interpreting results in ATXN1 phosphorylation studies.

How might targeting ATXN1 phosphorylation lead to therapeutic approaches for SCA1?

Understanding ATXN1 phosphorylation provides several potential therapeutic strategies for SCA1:

  • PKA inhibition: Since PKA appears to be the primary kinase responsible for ATXN1 phosphorylation at S776 in cerebellar Purkinje cells , selective PKA inhibitors could potentially reduce phosphorylation. By decreasing phosphorylation, ATXN1 stability might be reduced, potentially lowering levels of toxic expanded polyglutamine ATXN1.

  • Phosphatase activation: Enhancing the activity of phosphatases that remove the phosphate group from S776 could increase ATXN1 turnover and reduce toxicity by promoting protein degradation.

  • Disruption of phosphorylation-dependent interactions: Phosphorylation at S776 likely mediates specific protein-protein interactions. Compounds that interfere with these interactions could mitigate downstream pathological effects without broadly affecting phosphorylation mechanisms.

  • Combined approaches: Since phosphorylation affects multiple aspects of ATXN1 biology, including stability, localization, and interactions with other proteins, combination therapies targeting multiple aspects might be more effective than single-target approaches.

  • Biomarker applications: Phospho-ATXN1 (S776) levels could potentially serve as biomarkers for disease progression or treatment response, allowing for pharmacodynamic monitoring during clinical trials.

The development of these approaches faces challenges including achieving sufficient specificity to avoid off-target effects and ensuring adequate delivery to cerebellar Purkinje cells, the primary site of SCA1 pathology.

What methodologies can assess compounds affecting ATXN1 phosphorylation?

Researchers evaluating compounds that modulate ATXN1 phosphorylation can employ several complementary methodological approaches:

  • In vitro screening systems:

    • Cerebellar extract-based phosphorylation assays using recombinant GST-ATXN1 substrates

    • Purified PKA-based assays to test direct effects on kinase activity

    • High-throughput screening platforms with phospho-specific antibody readouts

  • Cellular validation approaches:

    • Western blot analysis of phospho-ATXN1 (S776) levels in treated cells

    • Immunofluorescence to assess effects on subcellular localization

    • Protein stability assays to determine impact on ATXN1 turnover

    • Cell models expressing either wild-type (30Q) or expanded (82Q) ATXN1

  • In vivo evaluation:

    • Transgenic mouse models (e.g., ATXN1[82Q]-S776 mice)

    • Behavioral assessments (e.g., rotarod performance)

    • Biochemical analysis of tissue extracts for phospho-ATXN1 levels

    • Histological examination of cerebellar Purkinje cells

  • Target validation approaches:

    • Comparison with genetic models (e.g., ATXN1-A776 phospho-resistant mutations)

    • Epistasis experiments with kinase modulators and phosphatase activators

    • Evaluation of effects on downstream pathways affected by phosphorylated ATXN1

These methodologies provide a comprehensive framework for evaluating compound effects on ATXN1 phosphorylation from initial screening through in vivo validation.

How can phospho-specific antibodies be used to monitor disease progression or treatment response?

Phospho-ATXN1 (Ser776) antibodies offer valuable tools for monitoring disease mechanisms and therapeutic interventions:

  • Biomarker development:

    • Quantitative measurement of phospho-ATXN1 (S776) levels in accessible biofluids or tissues could potentially track disease progression

    • Longitudinal monitoring during clinical interventions might provide pharmacodynamic evidence of target engagement

  • Mechanism of action studies:

    • Determining whether experimental therapeutics affect phosphorylation status, protein levels, or downstream effects

    • Differentiating between compounds that directly affect phosphorylation versus those that act through other mechanisms

  • Patient stratification:

    • Potentially identifying patient subgroups with different phosphorylation profiles who might respond differently to specific therapeutic approaches

    • Correlating phosphorylation levels with disease severity or progression rates

  • Methodological approaches:

    • ELISA-based quantification using phospho-specific antibodies

    • Immunohistochemical analysis of post-mortem samples or animal models

    • Multiplexed assays combining phospho-ATXN1 with other disease-relevant biomarkers

  • Technical considerations:

    • Sample preparation standardization is crucial for reliable quantification

    • Inclusion of appropriate controls for antibody specificity validation

    • Development of robust normalization strategies to account for total ATXN1 levels

These applications demonstrate the translational potential of phospho-specific antibodies beyond basic research contexts.

What controls should be included when studying ATXN1 phosphorylation?

A comprehensive experimental design for studying ATXN1 phosphorylation should include several types of controls:

  • Antibody specificity controls:

    • Phospho-peptide blocking: Pre-incubating antibodies with the phosphopeptide used as immunogen should eliminate specific binding

    • Non-phosphorylated control peptides: Should not affect antibody binding

    • Secondary antibody-only controls: To assess background signal

  • Genetic controls:

    • Phospho-resistant mutants: ATXN1-A776 constructs serve as negative controls that cannot be phosphorylated at position 776

    • Wild-type ATXN1-S776 constructs: Positive controls that can be phosphorylated

    • Varying polyglutamine lengths: Comparing ATXN1[30Q] versus ATXN1[82Q] can reveal polyglutamine length effects

  • Enzymatic controls:

    • Phosphatase treatment: Samples treated with phosphatases should show reduced or eliminated phospho-specific signals

    • Kinase inhibitors: PKA inhibitors like staurosporine should reduce phosphorylation in assay systems

    • Kinase activators: Agents that increase PKA activity can serve as positive controls

  • Treatment controls:

    • Known modulators: Adriamycin treatment (0.5μM, 5h) has been shown to affect ATXN1 phosphorylation in certain cell types

    • Time course analyses: To capture dynamic phosphorylation events

    • Dose-response studies: To establish optimal treatment parameters

These controls help establish the specificity of detection methods and provide context for interpreting experimental results in phosphorylation studies.

What are optimal methods for subcellular fractionation when studying ATXN1 phosphorylation?

Subcellular fractionation is particularly important for ATXN1 phosphorylation studies given the finding that kinase activity is enriched in the cytoplasmic fraction while phosphorylated ATXN1 accumulates predominantly in the nuclear fraction . Optimal methods include:

  • Fractionation approach:

    • Differential centrifugation: Sequential centrifugation steps at increasing speeds can separate nuclei from cytoplasmic components

    • Density gradient separation: For higher purity fractions when needed

    • Commercial kits: Several validated nuclear/cytoplasmic fractionation kits are available

  • Critical considerations:

    • Phosphatase inhibitors: Must be included in all buffers to preserve phosphorylation status

    • Protease inhibitors: Essential to prevent protein degradation

    • Temperature control: Performing all steps at 4°C helps maintain protein integrity and phosphorylation

    • Buffer composition: Hypotonic buffers for cell swelling followed by mechanical disruption are often effective

  • Validation of fraction purity:

    • Western blotting for compartment-specific markers:

      • Nuclear markers: Lamin B, histone H3

      • Cytoplasmic markers: GAPDH, α-tubulin

    • Microscopic examination: To confirm nuclear integrity during isolation

  • Application to ATXN1 studies:

    • Analysis has shown that pS776-endogenous ATXN1 is enriched in nuclear fractions

    • Kinase activity assays using fractions have demonstrated that S776 kinase activity is predominantly found in cytoplasmic fractions

    • Combining fractionation with immunoprecipitation can enrich for specific ATXN1 populations

These approaches allow researchers to study the subcellular compartmentalization of ATXN1 phosphorylation processes and potentially identify the site of initial phosphorylation versus the site of phosphorylated protein accumulation.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.