ATXN2 Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to ATXN2 Antibody

ATXN2 antibodies are specialized immunological tools designed to detect and study ataxin-2 (ATXN2), a eukaryotic RNA-binding protein implicated in mRNA metabolism and neurodegenerative diseases. These antibodies enable researchers to investigate ATXN2's molecular functions, isoforms, and pathological roles in disorders such as spinocerebellar ataxia type 2 (SCA2) and amyotrophic lateral sclerosis (ALS) .

Protein Structure

ATXN2 contains:

  • Two LSm domains for RNA binding.

  • A PAM2 motif for interaction with poly(A)-binding protein (PABP).

  • An N-terminal polyglutamine (polyQ) tract (14–31 residues in healthy individuals), which expands pathologically in SCA2 (32–200 residues) .

PropertyDetails
Molecular weight12–140 kDa, depending on isoform
IsoformsMultiple isoforms with varying lengths and domain compositions
Subcellular localizationCytoplasmic, associated with Golgi apparatus and stress granules
Tissue specificityHighly expressed in CNS; also detected in liver and gallbladder

Research Applications of ATXN2 Antibodies

ATXN2 antibodies are critical for:

  • Western blot (WB): Detects full-length ATXN2 (~140–150 kDa) and proteolytic fragments (e.g., 42 kDa polyQ-containing fragment) .

  • Immunohistochemistry (IHC): Localizes ATXN2 in tissues such as brain, breast cancer, and gallbladder .

  • Flow cytometry: Quantifies ATXN2 expression in cell lines (e.g., RT4 cells) .

  • TR-FRET assays: Measures soluble polyQ-expanded ATXN2 in human biomaterials (e.g., cerebrospinal fluid) .

Validated Antibody Examples

AntibodyHostApplicationsKey FindingsSource
21776-1-AP (Proteintech)RabbitWB, IHC, TR-FRETDetects polyQ-expanded ATXN2 in ALS and SCA2 models Proteintech
PA2098 (Boster Bio)RabbitWB, IHC, IF, Flow CytometryIdentifies ATXN2 at ~140 kDa in human/mouse tissues Boster Bio

Neurodegenerative Diseases

  • SCA2: Caused by polyQ expansions (>33 repeats) in ATXN2, leading to cerebellar degeneration .

  • ALS: Intermediate polyQ expansions (27–33 repeats) increase disease risk by promoting TDP-43 aggregation .

Therapeutic Targeting

  • Antisense oligonucleotides (ASOs): Reduce ATXN2 mRNA levels in ALS models, improving survival and motor function .

  • TR-FRET biomarkers: Enable quantification of soluble polyQ-expanded ATXN2 for clinical trials .

Proteolytic Cleavage of ATXN2

  • Full-length ATXN2 undergoes cleavage into:

    • N-terminal fragment (42 kDa): Contains polyQ tract and aggregates in SCA2 brains .

    • C-terminal fragment (70 kDa): Lacks polyQ tract and shows no disease-specific changes .

  • Proteolysis contributes to SCA2 pathology by releasing toxic polyQ fragments .

ATXN2 in Stress Granules

  • ATXN2 regulates stress granule dynamics, influencing mRNA stability and TDP-43 mislocalization .

  • Depleting ATXN2 with ASOs reverses TDP-43 pathology in ALS neurons .

Challenges and Future Directions

  • Standardization: Variability in ATXN2 isoform detection complicates biomarker studies .

  • Therapeutic development: Phase I trials (e.g., ASO BIIB105) aim to validate ATXN2 as a druggable target .

Product Specs

Buffer
PBS with 0.02% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze/thaw cycles.
Lead Time
Typically, we can ship your orders within 1-3 business days after receiving them. Delivery time may vary depending on the purchase method or location. For specific delivery information, please consult your local distributors.
Synonyms
ASL13 antibody; Ataxin 2 antibody; Ataxin-2 antibody; ATX2_HUMAN antibody; Atxn2 antibody; Olivopontocerebellar ataxia 2; autosomal dominant antibody; SCA2 antibody; Spinocerebellar ataxia type 2 protein antibody; TNRC13 antibody; Trinucleotide repeat containing 13 antibody; Trinucleotide repeat containing gene 13 protein antibody; Trinucleotide repeat-containing gene 13 protein antibody
Target Names
ATXN2
Uniprot No.

Target Background

Function
ATXN2 Antibody is involved in EGFR trafficking, acting as a negative regulator of endocytic EGFR internalization at the plasma membrane.
Gene References Into Functions
  1. STAU1 is recruited to mutant ATXN2 aggregates in spinocerebellar ataxia type 2 fibroblasts. PMID: 30194296
  2. Intermediate-length ATXN2 repeat expansions might be a risk factor in Korean patients with ALS. PMID: 29665996
  3. A novel variant in ATXN2 was identified in a Chinese population and linked to age of onset in Machado-Joseph disease. PMID: 27452601
  4. Correlations were found, including that between dystonia and CAG expansion. Notably, there was an association between cognitive decline and the variant G at mitochondrial polymorphism A10398G, previously associated with earlier ages at onset in SCA2. PMID: 28648514
  5. SCA2 should be considered as a potential cause of typical Parkinson's disease phenotype even in the absence of cerebellar ataxia. PMID: 28462804
  6. This is the first study suggesting a possible association between the (CAG)n polymorphism of the ATXN2 gene and the development of pure type 2 diabetes in a low-resource population. Long normal alleles of the VNTR are risk factors for pure type 2 diabetes in the analyzed Mexican population. PMID: 28076580
  7. Deleterious non-synonymous single nucleotide polymorphisms in the ATXN2 gene are associated with protein instability and conformational changes resulting in spinocerebellar ataxia. PMID: 28612427
  8. Intermediate expansions of the CAG repeat in ATXN2 are associated with amyotrophic lateral sclerosis. They are primarily associated with TDP-43 proteinopathy, but not with 1C2-positive polyglutamine inclusions. PMID: 26095883
  9. The findings of this study suggest that ATXN2 may modify the known PINK1 roles for mitochondrial quality control and autophagy during cell stress. PMID: 27597528
  10. The conclusion of this study suggests that the transcriptome data do not exclude the role of ATXN2 mutated alleles in Parkinson disease. However, its decreased protein expression in both SCA2c and SCA2p patients suggests a potential involvement of this gene in Parkinson disease. PMID: 27663142
  11. C9orf72 and ATXN2 repeat expansions cause ataxia, dementia, and parkinsonism in a Guyana family. PMID: 28124431
  12. Intermediate length repeat expansions of CAG (polyQ) repeat in the ATXN2 gene have also been reported to increase the risk of developing ALS. PMID: 28527524
  13. Intermediate ATXN2 trinucleotide repeat expansion in ALS does not predict age of onset but does predict disease risk. PMID: 28017481
  14. While depletion of C9ORF72 alone has a partial deleterious effect on neuron survival, it synergizes with the toxicity of Ataxin-2 carrying intermediate length of polyglutamine expansions to induce motor neuron dysfunction and neuronal cell death. PMID: 27103069
  15. ATXN2-AS, a gene antisense to ATXN2, has a role in SCA2 and possibly ALS pathogenesis. PMID: 27531668
  16. It is not a regulatory gene in Italian amyotrophic lateral sclerosis patients with C9ORF72 GGGGCC expansion. PMID: 26733254
  17. Selective loss of Purkinje cells in the cerebellar vermis of amyotrophic lateral sclerosis cases with intermediate repeat expansions in the ATXN2 gene. PMID: 26599997
  18. A meta-analysis of the top SNPs identified three new associated loci in primary open angle glaucoma: TXNRD2, ATXN2, and FOXC1. PMID: 26752265
  19. Data suggest that the spinocerebellar ataxia 2 protein (ATXN2, SCA2) CAG/CAA repeat expansion may play a significant role in the phenotypic variability of Parkinson's disease. PMID: 26663046
  20. ATXN2 is a modifier of phenotype in ALS patients of Sardinian ancestry. PMID: 26208502
  21. FBXW8 and PARK2 are sequestrated into insolubility by ATXN2 PolyQ expansions, but only FBXW8 expression is dysregulated. PMID: 25790475
  22. This is the first description of a family with two SCA mutations, with affected subjects having a combined SCA2 and SCA10 phenotype. PMID: 25630585
  23. ATXN2 intermediate-length polyglutamine expansions greater than 24 and 27 repeats were associated with sporadic ALS. PMID: 25457026
  24. ATXN2 CAG expansion is the sole causative mutation responsible for the parkinsonian phenotype of spinocerebellar ataxia-2. PMID: 25189117
  25. Results indicate the presence of intermediate CAG repeat expansion in the ATXN2 gene is a specific genetic risk factor for amyotrophic lateral sclerosis [review, meta-anlysis]. PMID: 25148523
  26. Review of the role of epigenetics and the ATXN2 gene in spinocerebellar ataxia 2 and amyotrophic lateral sclerosis. PMID: 24485162
  27. This study demonstrated that ATXN2 polyQ intermediate-length repeat is a modifier of ALS survival in the Italian population. PMID: 25527265
  28. The data of this study showed that a total of 64.3% of familial and 27.8% of sporadic subjects carried potentially pathogenic novel or rare coding variants identified by sequencing or an expanded repeat in C9ORF72 or ATXN2. PMID: 25382069
  29. This meta-analysis calculates the association between ATXN2 CAG repeat alleles and increased risk of ALS across multiple ethnic groups. PMID: 25285812
  30. Intermediate ATXN2 repeat lengths may render C9ORF72 expansion carriers more susceptible to developing motorneuron disease. PMID: 24866401
  31. Evidence supports the hypothesis of large normal alleles being a reservoir of expanded alleles in SCA2, related to autosomal dominant cerebellar ataxias. PMID: 23865735
  32. The ATXN2 gene may confer vulnerability for Schizophrenia. PMID: 24333172
  33. ATXN2 intermediary repeat length is a strong risk factor for amyotrophic lateral sclerosis (ALS) and ALS-frontotemporal dementia [FTD-ALS]. It is proposed that ATXN2 polyQ expansions could act as a modifier of the FTD phenotype in the presence of C9orf72 repeat expansion, leading to the development of clinical signs featuring both FTD and ALS. PMID: 25098532
  34. Cdk5 controls the abundance of both normal and polyQ-expanded ataxin-2 protein in neurons. PMID: 24486837
  35. Ataxin-2 is an RNA-binding protein that targets cis-regulatory elements in 3' UTRs to stabilize a subset of mRNAs and increase protein expression. PMID: 24954906
  36. Couples with no family history of SCA2 may have a greater than 0% risk of having an affected offspring. Similarly, couples in which there is both an expanded and a large normal allele may have a recurrence risk greater than 50%. PMID: 23813298
  37. The pathological expansions (greater than 34 repeats) of a CAG repeat in ATXN2, which encodes a polyglutamine tract in ataxin-2, cause spinocerebellar ataxia type 2. Intermediate-length expansions were reported to contribute to susceptibility to ALS. PMID: 24085347
  38. This study presented that early features of SCA2 are detectable before the onset of the cerebellar syndrome and are associated with expanded CAG repeats and the time to onset of cerebellar syndrome. PMID: 24657153
  39. The association of de novo mutations in ATXN2 with autosomal dominant amyotrophic lateral sclerosis. PMID: 23936447
  40. Thrombotic antiphospholipid syndrome shows strong haplotypic association with the SH2B3-ATXN2 locus. PMID: 23844121
  41. Our data indicate that, for ALS patients from mainland China, intermediate CAG repeat expansions in ATXN2 increase the risk of amyotrophic lateral sclerosis (ALS) but have no effect on disease phenotype. PMID: 23635656
  42. Data indicate that the expanded CAG repeat varied between 32 and 79 with a mean of 41.4 +/- 5.7 units. PMID: 22758789
  43. Golgi fragmentation was enhanced, and the early stages of apoptosis were triggered when ataxin-2 Q31 was co-expressed with mutant FUS. PMID: 23172909
  44. Ataxin 2 and ataxin 2-like have functional overlap, with ataxin-2-like having a role in the regulation of stress granules and processing bodies. PMID: 23209657
  45. Observations suggest that genotyping of SNPs at this locus may be useful for studying ALS risk in a high percentage of individuals, and that ATXN2 and SH2B3 variants may interact in modulating the disease pathway. PMID: 22916186
  46. ETS1 regulates the expression of ATXN2. PMID: 22914732
  47. Ataxin-2 has roles in pathological cascades mediated by TAR DNA-binding protein 43 (TDP-43) and Fused in Sarcoma (FUS). PMID: 23048034
  48. This study demonstrated that ATXN2 with intermediate-length CAG/CAA repeats does not appear to be a risk factor in hereditary spastic paraplegia. PMID: 22868089
  49. ATXN2 associated polyglutamine amplification is specific to the amyotrophic lateral sclerosis-end of the frontotemporal lobar degeneration-amyotrophic lateral sclerosis disease spectrum. PMID: 22035589
  50. Our findings indicate that only ATAXIN-2 alleles with >/= 31 CAG may represent low-penetrance disease/susceptibility alleles associated with variable neurodegenerative phenotypes, including cerebellar ataxia, parkinsonism, and ALS. PMID: 22425256

Show More

Hide All

Database Links

HGNC: 10555

OMIM: 183090

KEGG: hsa:6311

STRING: 9606.ENSP00000366843

UniGene: Hs.732512

Involvement In Disease
Spinocerebellar ataxia 2 (SCA2); Amyotrophic lateral sclerosis 13 (ALS13)
Protein Families
Ataxin-2 family
Subcellular Location
Cytoplasm.
Tissue Specificity
Expressed in the brain, heart, liver, skeletal muscle, pancreas and placenta. Isoform 1 is predominant in the brain and spinal cord. Isoform 4 is more abundant in the cerebellum. In the brain, broadly expressed in the amygdala, caudate nucleus, corpus cal

Q&A

What epitopes do commercial ATXN2 antibodies typically target?

Commercial ATXN2 antibodies generally target either N-terminal or C-terminal regions of the protein. C-terminal antibodies (often referred to as C-ATXN2) are most common, with several validated options available across multiple suppliers . N-terminal antibodies are less common, and researchers often use epitope-tagging approaches (such as HA-tagging) when studying N-terminal fragments .

Some key epitope regions include:

  • N-terminal regions upstream of the polyQ domain

  • The polyQ domain itself (specifically targeted by antibodies like 1C2 that detect expanded polyQ tracts)

  • Conserved domains such as the Lsm or LsmAD domains

  • C-terminal regions containing the PAM2 domain

When selecting an antibody, consider which region of ATXN2 is most relevant to your research question and whether you need to distinguish between different isoforms or cleavage products .

How can I validate the specificity of an ATXN2 antibody?

Validating ATXN2 antibody specificity requires multiple complementary approaches:

  • RNA interference validation: Create cell lines expressing different shRNAs targeting ATXN2 mRNA. A specific antibody should show reduced signal intensity in Western blots of these knockdown lines compared to controls, while maintaining consistent detection of control proteins like GAPDH .

  • Overexpression testing: Express tagged versions of ATXN2 (e.g., HA-tagged ATXN2) and confirm co-detection with both the tag-specific antibody and your ATXN2 antibody .

  • Immunoprecipitation: Perform IP with one antibody and detection with another targeting a different epitope to confirm specificity .

  • Comparative analysis with multiple antibodies: Use multiple antibodies targeting different ATXN2 epitopes to verify consistent protein detection patterns .

  • Negative controls: Include tissues or cell lines from ATXN2 knockout models when available .

Which applications are ATXN2 antibodies validated for?

Most commercial ATXN2 antibodies have been validated for multiple applications:

ApplicationValidated UsesSpecial Considerations
Western Blot (WB)Detection of full-length ATXN2 and cleavage productsRequires careful sample preparation to preserve protein integrity
Immunohistochemistry (IHC)Localization in tissue sectionsMay require antigen retrieval optimization
Immunocytochemistry (ICC)Subcellular localizationFixation method affects epitope accessibility
Immunoprecipitation (IP)Protein-protein interaction studiesBuffer optimization important for maintaining interactions
ELISAQuantitative detectionLimited commercial options available
TR-FRETSensitive detection of polyQ-expanded ATXN2Requires specialized fluorophore-conjugated antibodies

Multiple studies confirm Western blot and immunohistochemistry as the most reliable applications, with most antibodies detecting the expected ~145 kDa and ~180 kDa bands corresponding to different ATXN2 isoforms or cleavage products .

How can I detect polyQ-expanded ATXN2 versus normal ATXN2?

Detecting polyQ-expanded ATXN2 requires specialized approaches:

  • PolyQ-specific antibodies: The 1C2 antibody (clone 5TF1-1C2) specifically recognizes expanded polyQ tracts (typically >37Q) but not normal-length polyQ domains. This allows selective detection of mutant ATXN2 .

  • TR-FRET immunoassay: This highly sensitive approach uses two antibodies - one ATXN2-specific antibody labeled with a donor fluorophore (Tb) and one polyQ-specific antibody labeled with an acceptor fluorophore (D2). Energy transfer only occurs when both antibodies bind in close proximity, allowing selective detection of polyQ-expanded ATXN2 .

  • Gel mobility shift analysis: PolyQ-expanded ATXN2 migrates more slowly in SDS-PAGE compared to normal ATXN2, creating a detectable mobility shift that correlates with polyQ length. This approach requires high-resolution gels and optimized running conditions .

  • Combination approach: For highest specificity, combine a general ATXN2 antibody with a polyQ-specific antibody (like 1C2) on parallel blots of the same samples .

The TR-FRET method offers particular advantages for quantitative analysis, with sensitivity sufficient to detect small changes in protein expression following treatments like siRNA knockdown or starvation .

How can I establish a TR-FRET-based immunoassay for quantifying polyQ-expanded ATXN2?

Establishing a TR-FRET immunoassay for polyQ-expanded ATXN2 requires several optimization steps:

  • Antibody selection and labeling:

    • Select two ATXN2-specific antibodies (e.g., ataxin-2 polyclonal antibody 21776-1-AP from Proteintech and purified mouse anti-ataxin-2 monoclonal antibody AB_398900 from BD)

    • Label them with donor fluorophore Tb

    • Select polyQ-specific antibodies (e.g., clone MW1 AB_528290 or clone 5TF1-1C2 MAB1574)

    • Label them with acceptor fluorophore D2

  • Buffer optimization:

    • Test different buffer conditions to maximize signal-to-background ratio

    • Include protease inhibitors to prevent protein degradation

    • Optimize detergent concentrations to maintain protein solubility while preserving antibody binding

  • Assay validation:

    • Use recombinant ATXN2 with normal (22Q) and expanded (79Q) polyQ tracts as controls

    • Validate in cellular models (e.g., HEK293T cells transfected with ATXN2 constructs)

    • Test in patient-derived materials like iPSC-neurons

  • Sensitivity testing:

    • Determine lower limits of detection

    • Validate ability to detect changes following treatments (e.g., siRNA knockdown)

The established TR-FRET method has demonstrated sufficient sensitivity to detect small changes in ATXN2 expression and can be applied to various biological samples including cell lysates and potentially biofluids .

What techniques can detect ATXN2 N-terminal proteolytic fragments?

Detecting ATXN2 N-terminal fragments requires specialized techniques:

Remember that N-terminal ATXN2 fragments containing expanded polyQ tracts have been observed in brain extracts from SCA2 patients and may have pathological significance .

How can I analyze ATXN2 subcellular localization in neuronal models?

Analyzing ATXN2 subcellular localization requires careful experimental design:

  • Co-localization studies:

    • Combine ATXN2 antibodies with markers for specific subcellular compartments

    • Ribosomal protein S6 and poly-A binding protein 1 (translation machinery)

    • Calnexin (endoplasmic reticulum)

    • TIA-1 or G3BP1 (stress granules)

    • pTDP-43 (in disease models)

  • Fixation optimization:

    • Different fixation methods preserve different protein interactions

    • Paraformaldehyde fixation (4%, 10-15 minutes) works well for most applications

    • For stress granule co-localization, brief methanol fixation may better preserve RNA-protein complexes

  • Super-resolution microscopy:

    • Standard confocal microscopy may not resolve fine structures

    • STED or STORM microscopy provides enhanced resolution to distinguish ATXN2 within complex structures like stress granules

  • Live-cell imaging:

    • For dynamic processes like stress granule formation, use fluorescently-tagged ATXN2 constructs

    • Verify that tagging doesn't alter localization by comparing with immunostaining of endogenous ATXN2

  • Biochemical fractionation:

    • Complement imaging with subcellular fractionation (cytosolic, membrane, nuclear fractions)

    • Use Western blotting with ATXN2 antibodies to quantify distribution

Normal ATXN2 typically localizes to the cytoplasm and proximal dendrites in neurons, with strong association with protein synthesis machinery, while expanded ATXN2 may show altered localization patterns in disease models .

What are the best approaches for investigating ATXN2 in post-mortem human brain samples?

Working with post-mortem tissue requires specialized approaches:

  • Sample preparation:

    • Optimize tissue preservation - fresh frozen tissue generally yields better results than formalin-fixed paraffin-embedded tissue

    • For protein extraction, use buffers containing detergents like sarkosyl to solubilize membrane-associated proteins

    • Separate sarkosyl-soluble and sarkosyl-insoluble fractions to distinguish aggregated from soluble ATXN2

  • Immunohistochemistry protocol:

    • Use heat-mediated antigen retrieval (citrate buffer pH 6.0)

    • Block endogenous peroxidases and biotin

    • Include Sudan Black B treatment to reduce autofluorescence if using fluorescent detection

    • Use Tyramide Signal Amplification for weak signals

  • Regional analysis:

    • Focus on cerebellar Purkinje cells (primary pathology in SCA2)

    • Also examine hippocampal and cortical regions (relevant for TDP-43 pathology)

  • Co-localization studies:

    • In FTLD-TDP cases, examine co-localization with phosphorylated TDP-43 in neuronal cytoplasmic inclusions and dystrophic neurites

    • Use sequential double-labeling techniques to minimize cross-reactivity

  • Quantitative analysis:

    • Use semi-quantitative immunofluorescence analysis to measure ATXN2 levels

    • Compare protein levels between disease cases and controls using Western blot densitometry

Studies of FTLD-TDP cases have revealed colocalization of ATXN2 with phosphorylated TDP-43 in pathological inclusions and significant reduction of ATXN2 protein compared to controls, suggesting involvement in TDP-43 proteinopathies .

How do I distinguish between different ATXN2 isoforms and cleavage products?

Distinguishing between ATXN2 isoforms requires multiple analytical approaches:

  • Molecular weight analysis:

    • Full-length ATXN2 (from ATG1 start site): ~180 kDa apparent molecular weight

    • Shorter ATXN2 isoform (from ATG2 start site): ~145 kDa

    • N-terminal cleavage products with normal polyQ: ~27 kDa

    • N-terminal cleavage products with expanded polyQ: ~30 kDa

    • C-terminal fragments lacking polyQ domain: ~145 kDa or smaller

  • Epitope mapping:

    • Use N-terminal tag antibodies (e.g., HA) to detect N-terminal fragments

    • Use C-terminal ATXN2 antibodies to detect C-terminal fragments

    • Use polyQ-specific antibodies (1C2) to confirm polyQ presence in fragments

  • Expression constructs:

    • Express ATXN2 from specific start codons (ATG1 vs. ATG2)

    • Create deletion constructs to identify cleavage sites

    • Compare migration patterns on Western blots

  • Immunoprecipitation:

    • IP with N-terminal antibodies to determine which fragments remain associated after cleavage

    • IP with C-terminal antibodies to pull down C-terminal fragments

  • Gradient gel electrophoresis:

    • Use 4-20% gradient gels to resolve both high and low molecular weight species

    • Apply appropriate molecular weight markers spanning the full range

Research has shown that full-length ATXN2 migrates more slowly than predicted (~180 kDa vs. predicted ~140 kDa), while N-terminal cleavage products containing the polyQ domain show mobility differences that correlate with polyQ length .

What are the critical factors in developing PolyQ-specific detection of ATXN2?

Developing polyQ-specific detection systems requires attention to several critical factors:

  • Antibody selection:

    • The monoclonal antibody 1C2 (clone 5TF1-1C2, MAB1574) specifically recognizes expanded polyQ stretches (typically >37Q)

    • Alternative polyQ antibodies include clone MW1 (AB_528290)

    • Different polyQ antibodies may have different threshold sensitivities for polyQ length

  • Signal-to-noise optimization:

    • Include appropriate negative controls (cells expressing normal-length ATXN2)

    • Titrate antibody concentrations to minimize background

    • For TR-FRET assays, optimize donor/acceptor antibody ratios

  • Buffer conditions:

    • Detergent selection affects polyQ epitope accessibility

    • Salt concentration can influence antibody specificity

    • pH optimization may be necessary for maximum binding

  • PolyQ length considerations:

    • Establish detection thresholds for different polyQ lengths

    • Use constructs with varying polyQ lengths (e.g., 22Q, 39Q, 79Q) as standards

    • Account for potential CAA interruptions in the CAG repeat that may affect antibody binding

  • Protein conformation:

    • Native vs. denatured protein may affect polyQ accessibility

    • Include protein denaturants like SDS for Western blot applications

    • Consider native conditions for TR-FRET applications

TR-FRET-based methods combining ATXN2-specific and polyQ-specific antibodies offer particular advantages for sensitive and specific detection of expanded ATXN2, with demonstrated utility in monitoring treatment effects in cellular models .

How can I monitor changes in ATXN2 levels following experimental treatments?

Monitoring ATXN2 changes requires quantitative approaches:

  • Western blot quantification:

    • Use housekeeping proteins (β-actin, GAPDH) as loading controls

    • Apply densitometry analysis with appropriate software

    • Run standard curves using recombinant ATXN2 for absolute quantification

  • TR-FRET quantification:

    • Particularly useful for detecting small changes in polyQ-expanded ATXN2

    • Has been validated for monitoring siRNA knockdown effects

    • Can detect changes following cellular stresses like starvation

  • qPCR for transcript changes:

    • Design primers spanning exon-exon junctions

    • Include reference genes (GAPDH, ACTB)

    • Correlate with protein changes to identify post-transcriptional regulation

  • Immunocytochemistry with quantitative image analysis:

    • Use consistent acquisition parameters

    • Apply automated image analysis for unbiased quantification

    • Include nuclear or cytoskeletal markers for normalization

  • Experimental timeline considerations:

    • For siRNA experiments, 72-hour post-transfection timepoint has been validated

    • For starvation experiments, effects have been observed within 1-2 hours

The TR-FRET immunoassay has been validated as sufficiently sensitive to detect small changes in ATXN2 expression following treatments like siRNA knockdown or starvation, making it particularly valuable for therapeutic monitoring applications .

What are the key considerations when studying ATXN2 and TDP-43 interactions?

Studying ATXN2-TDP-43 interactions requires specialized approaches:

  • Co-immunoprecipitation strategy:

    • Use antibodies against native proteins or epitope tags

    • Include RNase treatment controls to distinguish RNA-dependent interactions

    • Use crosslinking approaches for transient interactions

  • Co-localization analysis:

    • In FTLD-TDP cases, focus on hippocampal regions

    • Look for colocalization in both neuronal cytoplasmic inclusions and dystrophic neurites

    • Use phospho-TDP-43 specific antibodies to identify pathological TDP-43

  • Genetic modification approaches:

    • Use ATXN2 knockdown to assess effects on TDP-43 aggregation

    • Compare normal vs. expanded polyQ ATXN2 effects on TDP-43 localization

    • Consider intermediate polyQ expansions relevant to ALS/FTLD

  • Stress response analysis:

    • Examine stress granule dynamics where both proteins localize

    • Apply stressors like arsenite, heat shock, or starvation

    • Monitor recovery phases to assess granule persistence

  • Biochemical fractionation:

    • Separate detergent-soluble and insoluble fractions

    • Compare distribution of both proteins across fractions

    • Assess changes in solubility following stress or in disease models

Research has shown reduced ATXN2 protein levels in FTLD-TDP cases compared to controls, with ATXN2 colocalizing with phosphorylated TDP-43 in pathological inclusions, suggesting a functional relationship relevant to disease mechanisms .

What controls are essential when studying ATXN2 proteolytic cleavage?

Studying ATXN2 proteolytic cleavage requires rigorous controls:

  • Construct controls:

    • Compare full-length (ATG1) vs. shortened (ATG2) ATXN2 constructs

    • Include both normal (22Q) and expanded (39Q/79Q) polyQ variants

    • Generate deletion constructs to map cleavage sites

  • Protease inhibitor controls:

    • Include protease inhibitor cocktails in some samples

    • Test specific inhibitors to identify protease classes involved

    • Compare fresh vs. delayed extraction to assess post-lysis artifacts

  • Fragment authentication:

    • Confirm N-terminal fragments using both tag antibodies and polyQ antibodies

    • Verify C-terminal fragments using C-terminal ATXN2 antibodies

    • Perform immunoprecipitation to confirm fragment identity

  • Heterologous protein controls:

    • Test putative cleavage sequences by inserting into heterologous proteins

    • Confirm sufficiency of identified sequences for directing proteolysis

    • Use irrelevant sequences as negative controls

  • In vivo relevance controls:

    • Compare cellular findings with patient tissue samples

    • Analyze both control and disease brain extracts

    • Consider post-mortem interval effects on proteolysis

Research has demonstrated that both normal and expanded polyQ ATXN2 undergo proteolytic cleavage, releasing polyQ-containing N-terminal fragments, with potential implications for SCA2 pathogenesis similar to other polyQ disorders .

How can ATXN2 antibodies be used to evaluate therapeutic efficacy in SCA2 models?

ATXN2 antibodies offer several approaches for therapeutic monitoring:

  • Protein lowering assessment:

    • Quantify ATXN2 reduction following ASO or siRNA treatments

    • Use TR-FRET assays for sensitive detection of polyQ-expanded ATXN2

    • Measure both mRNA and protein levels to assess correlation

  • Tissue-specific evaluation:

    • Assess treatment effects across different tissues (cerebellum, brainstem, spinal cord)

    • Use immunohistochemistry to evaluate regional differences in protein lowering

    • Compare effects in neurons vs. glial cells

  • Isoform-specific analysis:

    • Determine whether treatments affect all ATXN2 isoforms equally

    • Monitor both full-length protein and proteolytic fragments

    • Assess normal vs. expanded polyQ ATXN2 selectivity

  • Cellular phenotype correlation:

    • Relate ATXN2 reduction to cellular phenotypes (stress granule dynamics, translation efficiency)

    • Correlate with downstream pathway markers

    • Use proximity ligation assays to assess protein-protein interactions

  • Biofluid biomarker development:

    • Apply TR-FRET assays to CSF or plasma samples

    • Establish baseline measures and variation in patient cohorts

    • Track longitudinal changes during therapeutic interventions

The established TR-FRET immunoassay for polyQ-expanded ATXN2 has demonstrated sufficient sensitivity for therapeutic monitoring applications and has potential utility as a pharmacodynamic biomarker in clinical trials .

What are the challenges in developing ATXN2 as a biomarker for neurodegenerative diseases?

Developing ATXN2 as a biomarker faces several challenges:

  • Sample accessibility:

    • ATXN2 levels in accessible biofluids (blood, CSF) may not reflect brain levels

    • Optimization of extraction protocols for different sample types is needed

    • Blood-brain barrier limits CNS protein detection in peripheral samples

  • Assay sensitivity requirements:

    • Low abundance in biofluids requires highly sensitive detection methods

    • TR-FRET approaches offer promising sensitivity but need optimization for biofluids

    • Amplification methods may be necessary for reliable detection

  • Isoform complexity:

    • Multiple ATXN2 isoforms and cleavage products complicate interpretation

    • Need for assays that distinguish pathogenic from normal forms

    • Potential post-translational modifications affecting detection

  • Disease specificity considerations:

    • ATXN2 alterations occur in multiple disorders (SCA2, ALS, FTLD-TDP)

    • Need to establish disease-specific patterns of change

    • Requirement for differential diagnostic potential

  • Validation requirements:

    • Correlation with clinical measures and disease progression

    • Establishment of age and sex-specific reference ranges

    • Validation across multiple cohorts and centers

The development of TR-FRET-based immunoassays specific for polyQ-expanded ATXN2 represents a promising approach for biomarker development, with demonstrated sensitivity in cellular models, but additional validation in patient biofluids is still needed .

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.