TARDBP Antibody

Shipped with Ice Packs
In Stock

Description

Biological Function and Clinical Relevance of TDP-43

TDP-43, encoded by the TARDBP gene (UniProt ID Q13148), regulates transcription, mRNA splicing, and RNA stability . Its pathological aggregation is a hallmark of:

  • Amyotrophic lateral sclerosis (ALS)

  • Frontotemporal lobar degeneration (FTLD)

  • Alzheimer's disease
    In cancer biology, TDP-43 exhibits dual roles: promoting survival in glioblastomas and lung cancers while suppressing tumor growth in rhabdomyosarcomas through miRNA regulation .

Neurodegenerative Disease Biomarkers

  • Serum anti-TDP-43 autoantibodies show 85% elevation in ALS patients compared to controls (p < 0.001)

  • IHC staining patterns differentiate ALS/FTLD (cytoplasmic inclusions) from normal nuclear localization

Oncological Investigations

  • Pro-tumor effects:

    • Enhances autophagy in glioblastomas under nutrient stress

    • Promotes metastasis in melanoma via glucose metabolism modulation

  • Anti-tumor effects:

    • Suppresses rhabdomyosarcoma growth through miR-500a-3p regulation

Validation Studies and Performance Metrics

A 2023 systematic evaluation of 18 commercial antibodies identified key performers:

  • Western Blot: Clone 982022 detected 43 kDa band across species with KO validation

  • Immunofluorescence: BSB-166 showed nuclear/cytoplasmic localization in ≤3 µg/mL concentrations

  • Immunoprecipitation: 4R5L7 demonstrated efficient protein recovery in neural tissue lysates

Technical Considerations for Experimental Use

ParameterRecommendationRationale
Antigen Retrieval10 mM PBS pH 7.2 (microwave method)Optimal for paraffin-embedded tissues
Antibody Dilution1:1000 (WB), 1:100 (IHC)Validated in multi-laboratory studies
Control TissuesBreast, brain cortex, glioblastoma Ensure proper localization patterns

Product Specs

Buffer
PBS with 0.1% 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 of receipt. Delivery times may vary based on the purchasing method or location. Please consult your local distributors for specific delivery times.
Synonyms
ALS10 antibody; OTTHUMP00000002171 antibody; OTTHUMP00000002172 antibody; OTTHUMP00000002173 antibody; TADBP_HUMAN antibody; TAR DNA binding protein 43 antibody; TAR DNA binding protein antibody; TAR DNA-binding protein 43 antibody; TARDBP antibody; TDP 43 antibody; TDP-43 antibody; TDP43 antibody
Target Names
Uniprot No.

Target Background

Function
TARDBP is an RNA-binding protein that plays a crucial role in various stages of RNA biogenesis and processing. Its two RNA recognition motifs (RRM1 and RRM2) preferentially bind to GU-repeats on RNA molecules, primarily located within long introns and the 3'UTR of mRNAs. This binding activity regulates the splicing of numerous non-coding and protein-coding RNAs, including those encoding proteins involved in neuronal survival, as well as mRNAs associated with neurodegenerative diseases. TARDBP contributes to mitochondrial homeostasis by controlling the processing of mitochondrial transcripts. It also regulates mRNA stability by recruiting the CNOT7/CAF1 deadenylase to mRNA 3'UTR, leading to poly(A) tail deadenylation and subsequent shortening. In response to oxidative stress, TARDBP associates with stalled ribosomes located in stress granules (SGs), contributing to cell survival. It also participates in normal skeletal muscle formation and regeneration, forming cytoplasmic myo-granules and binding mRNAs encoding sarcomeric proteins. TARDBP plays a role in maintaining the periodicity of the circadian clock by stabilizing CRY1 and CRY2 proteins in a FBXL3-dependent manner. Furthermore, it negatively regulates the expression of CDK6 and regulates the expression of HDAC6, ATG7, and VCP in a PPIA/CYPA-dependent manner.
Gene References Into Functions
  1. TDP-43 deposition leads to targeted RNA instability in amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). PMID: 30030424
  2. Mutations in CHCHD10 contribute to cytoplasmic TDP-43 accumulation and synaptic integrity. PMID: 28585542
  3. Studies have confirmed the high expression of hTDP-43 in the central nervous system (CNS), increased microgliosis, and motor deficits, exhibiting prominent ALS/FTLD pathologies, such as cytoplasmic and insoluble TDP-43 in TAR6/6 mice. This model demonstrates both pathological TDP-43 expression and disease-relevant posttranslational changes. PMID: 29787578
  4. Research highlights the relevance of contact-independent cell-to-cell transfer of TDP-43 and SOD1 in ALS. PMID: 28711596
  5. Findings indicate that the phosphatase regulator, GADD34, also functions as a kinase scaffold in response to chronic oxidative stress and recruits CK1 and oxidized TDP-43 to facilitate its phosphorylation, as observed in TDP-43 proteinopathies. PMID: 29109149
  6. Impaired RNA metabolism, secondary to TDP-43 loss of function, has been identified as a potential pathological mechanism of HSPB8 toxicity, leading to muscle and nerve degeneration. PMID: 29029362
  7. The introduction of SOD1(G93A) and TDP43(A315T), established ALS-related mutations, altered the subcellular expression and localization of RNAs within neurons, demonstrating a spatial specificity to either the soma or the axon. This research provides the first combined inclusive profile of mRNA and miRNA expression in two ALS models at the subcellular level. PMID: 28300211
  8. These data provide structural insights into the established mechanistic role of the well-folded TDP-43 NTD in splicing and link this function to liquid-liquid phase separation. PMID: 29438978
  9. TDP43 alters most splicing events with splicing factor SRSF3 in triple-negative breast cancer. PMID: 29581274
  10. Within a cohort of patients with Motor Neuron Disease and Frontotemporal Dementia, one patient exhibited a mutation in the TARDBP gene. PMID: 29886477
  11. Studies have demonstrated TDP-43/pTDP-43 deposition in skin nerves in ALS patients. Although the mechanisms underlying TDP-43 in ALS are currently unknown, its detection is significant, and the deposition may occur not only in ALS but also during the aging process, as observed in this study. PMID: 29804146
  12. Both ALS and FTD patients presented with higher TDP-43 and tauT levels compared to the control group. The combination of biomarkers in the form of the TDP-43 x tauT / tauP-181 formula achieved the best discrimination between ALS or FTD and controls, with sensitivities and specificities greater than 0.8. PMID: 28848086
  13. TDP-43 turnover and toxicity are partially dependent on the endocytosis pathway. TDP-43 inhibits endocytosis and co-localizes strongly with endocytic proteins, including in ALS patient tissue. PMID: 29233983
  14. A more selective group of neurons appears to be affected in frontotemporal lobar degeneration (FTLD)-TDP and FTLD-FUS than in FTLD-tau. PMID: 28984110
  15. A study found a high frequency of the TARDBP p.M337 V mutation in familial ALS in south-eastern China. Patients with TARDBP-linked ALS exhibited a benign disease course and prolonged survival. PMID: 29621978
  16. Two cases of apparently sporadic ALS associated with mutations in the SOD1 and TARDP genes were described. PMID: 27494151
  17. Research indicates that there are at least two common patterns of TDP-43 and tau protein misfolding during human brain aging. In patients without substantial Alzheimer's disease pathology, cerebral age-related TDP-43 with sclerosis (CARTS) cases tend to exhibit tau neurofibrillary tangles in the hippocampal dentate granule neurons, potentially serving as an indicator of CARTS. PMID: 28281308
  18. Depletion of TAF15, FUS, and TDP-43 in human-induced pluripotent stem cell-derived motor neurons affects different genes. PMID: 27378374
  19. TDP-43 mislocalization into axons precedes cell death in cortical neurons, and the expression of either wild-type or mutant TDP-43 constructs in vitro impairs cytoskeletal structure and function. PMID: 29787572
  20. TDP-43 impairs the induction of multiple key stress genes necessary for disease protection by reducing the recruitment of the chromatin remodeler Chd1 to chromatin. PMID: 29153328
  21. A mutation in the TARDBP gene has been linked to ALS. PMID: 29478603
  22. Cytoplasmic TDP-43 mislocalization and aggregation are characteristic pathological features of ALS and FTD. TDP-43 is an RNA-binding protein with a prion-like domain that promotes TDP-43 misfolding. [review] PMID: 27920024
  23. SOD1 mutations were present in 20% of familial ALS patients and 1.9% of sporadic ALS patients, while FUS mutations were responsible for 13.3% of familial ALS cases, and TARDBP mutations were rare in both familial and sporadic ALS cases. PMID: 27604643
  24. An alpha-helical component in the center (residues 320-340) of the C-terminal domain is associated with the protein's self-association and LLPS. Systematic analysis of ALS-related TDP-43 mutants (G298S, M337V, and Q331K) under different buffer conditions and temperatures demonstrates that this phase separation is driven by hydrophobic interactions but inhibited by electrostatic repulsion. PMID: 28988034
  25. This study did not demonstrate oxidative phosphorylation defects in TDP-43 mutants. PMID: 28482850
  26. Both FUS and TDP43 colocalize with active RNA polymerase II at sites of DNA damage alongside the DNA damage repair protein, BRCA1, and FUS and TDP43 participate in the prevention or repair of R loop-associated DNA damage, a manifestation of aberrant transcription and/or RNA processing. PMID: 27849576
  27. The data from this study suggest that TDP-43 pathology is associated with age and exacerbated by the presence of concomitant Alzheimer's disease pathology. PMID: 27495267
  28. The A382T mutation in TARDBP caused a reduction in the ability of cells to respond to stress through loss of TDP-43 function in stress granule nucleation. The pathogenetic action observed in this model does not appear to be mediated by changes in the localization of the TDP-43 protein. PMID: 28172957
  29. TDP-43 competes with other splicing factors for binding to cryptic exons and can repress cryptic exon inclusion. PMID: 28549443
  30. This study shows that TDP-43 is deposited in the olfactory bulb in Alzheimer's disease, albeit at a low frequency. The deposition appears to be a late occurrence compared to TDP-43 deposition in other brain regions. PMID: 26810591
  31. ALS-mutant linked TDP-43 mutations expressed at moderate levels, mimicking endogenous TDP-43, also cause toxicity in a non-cell autonomous manner. Eliminating mutant TDP-43Q331K synthesis in a proportion of motor neurons delayed disease onset, reduced aberrant nuclear morphology in those neurons at early disease stages, and nearly eliminated age-dependent accelerated death of those motor neurons. PMID: 28357566
  32. This study reports that cryptic exon incorporation occurred not only in Alzheimer's disease brains exhibiting TDP-43 pathology but also in neurons lacking cytoplasmic inclusions but exhibiting nuclear clearance of TDP-43. PMID: 28332094
  33. Acetylation of the protein triggers TDP-43 pathology in cultured cells and mouse skeletal muscle, which can be cleared through an HSF1-dependent chaperone mechanism that disaggregates the protein. PMID: 28724966
  34. These studies have shown that physiological oligomerization of TDP-43 is mediated through its N-terminal domain, which forms functional and dynamic oligomers that antagonize pathologic aggregation. PMID: 28663553
  35. Expression of PFN1 mutants induces accumulation of TDP-43 and promotes the conversion of normal TDP-43 into an abnormal form. These findings provide new insight into the mechanisms of TDP-43 proteinopathies and other diseases associated with amyloid-like protein deposition. PMID: 27432186
  36. This study reports the altered expression and/or mislocalization of the TAR-DNA binding protein 43 (TDP-43) in both Niemann-Pick disease type C mouse and in a human neuronal model of the disease. Results underscore the importance of TDP-43 in neurodegenerative disease and further highlight the need to prioritize targeting this protein to develop novel therapeutic strategies. PMID: 27193329
  37. This study demonstrated that increased rates of TDP-43-associated hippocampal atrophy might occur at least 10 years before death in patients with Alzheimer's disease. PMID: 28919059
  38. Authors observed impaired levels of glutathione (downstream Nrf2 antioxidant) in TDP-43M337V patient fibroblasts and astrocyte cultures from TDP-43Q331K mice, indicative of elevated oxidative stress and failure of some upregulated antioxidant genes to be translated into protein. PMID: 28334913
  39. Removing the human orthologs of Hrb27c (DAZAP1) in human neuronal cell lines can correct several pre-mRNA splicing events altered by TDP-43 depletion. PMID: 28575377
  40. TDP-43 suppressed tau expression by promoting its mRNA instability through the UG repeats of its 3-UTR. The C-terminal region of TDP-43 was required for this function. The level of TDP-43, which is decreased in AD brains, was found to correlate negatively with the tau level in the human brain. PMID: 28335005
  41. Amyotrophic lateral sclerosis mutations disrupt phase separation mediated by alpha-helical structure in the TDP-43 low-complexity C-terminal domain. PMID: 27545621
  42. Cytoplasmic TDP-43 aggregate formation was demonstrated in neuronal and glial cells following adenoviral transduction of WT and CTF TDP-43 under MG-132 treatment. These TDP-43 aggregates were phosphorylated and ubiquitinated and consisted of electron-dense granules. PMID: 28599005
  43. This research emphasizes the importance of distinguishing cerebral age-related TDP-43 with sclerosis from late-onset frontotemporal lobar degeneration with TDP-43 pathology and from advanced Alzheimer's disease with TDP-43 pathology. PMID: 28467211
  44. Mutant and wild-type human TDP-43 replacing the endogenous Drosophila gene reveals phosphorylation and ubiquitination in mutant lines without viability or lifespan defects. PMID: 28686708
  45. This study establishes a functional/physical partnership between FMRP and TDP-43, mechanistically linking several neurodevelopmental disorders and neurodegenerative diseases. PMID: 27518042
  46. By silencing TDP-43, authors observed significant inhibition of cell proliferation and metastasis in A375 and WM451 cells. TDP-43 knockdown suppressed glucose transporter type-4 (GLUT4) expression and reduced glucose uptake. PMID: 27786596
  47. This study, based on 15 cases staged for pTDP-43 pathology, reports that pathologically altered TDP-43 in Betz cells reacts differently than in bulbar or spinal alpha-motoneurons. The major differences between the two types of histological profiles are discussed in the context of their potential consequences and implications for the potential further progression or spread of the pTDP-43 lesions. PMID: 27757524
  48. This study has shown that TDP-43-positive cytoplasmic inclusions were frequently found in the amygdala of pathologically and genetically confirmed cases of Frontotemporal Lobar Degeneration and Motor Neuron Disease. PMID: 28859337
  49. Results indicated that a range of disease-specific TDP-43 variants are generated in ALS patients, with different variants being generated in sporadic and familial cases. PMID: 28122516
  50. There is heterogeneous structural reorganization and decreased stability of the truncated RRM2 domain. PMID: 28793209

Show More

Hide All

Database Links

HGNC: 11571

OMIM: 605078

KEGG: hsa:23435

STRING: 9606.ENSP00000240185

UniGene: Hs.300624

Involvement In Disease
Amyotrophic lateral sclerosis 10 (ALS10)
Subcellular Location
Nucleus. Cytoplasm. Cytoplasm, Stress granule.
Tissue Specificity
Ubiquitously expressed. In particular, expression is high in pancreas, placenta, lung, genital tract and spleen.

Q&A

What is TARDBP and why is it important in research?

TARDBP, also known as TDP-43, is an RNA and DNA binding protein belonging to the heterogeneous nuclear ribonucleoprotein (hnRNP) family. It contains two RNA recognition motif (RRM) domains that are crucial for RNA processing and regulation. TARDBP is ubiquitously expressed throughout the body, with highest expression levels in the placenta, lung, pancreas, spleen, and genital tract. The protein plays vital roles in several cellular processes including binding to TAR DNA sequence motifs of HIV where it functions as a transcriptional repressor inhibiting HIV-1 transcription. Additionally, TARDBP is involved in splicing of the cystic fibrosis transmembrane conductance regulator (CFTR) gene, highlighting its importance in maintaining proper cellular function . Most notably, TARDBP has gained significant attention in research due to its association with neurodegenerative diseases as it is a major component of ubiquitin-positive inclusion bodies found in conditions like ALS and FTLD .

What types of TARDBP antibodies are available for research applications?

TARDBP antibodies are available in several formats to accommodate different experimental needs. These include mouse monoclonal antibodies and rabbit polyclonal antibodies that can detect TARDBP protein from various species including human, mouse, and rat samples. The antibodies are available in both non-conjugated forms and various conjugated formats including agarose, horseradish peroxidase (HRP), phycoerythrin (PE), fluorescein isothiocyanate (FITC), and multiple Alexa Fluor® conjugates . Specific examples include the E-10 mouse monoclonal IgG2a kappa light chain antibody (sc-376311) which detects TARDBP across multiple species, and clone BSB-166, another mouse monoclonal antibody . The diversity of available antibodies allows researchers to select the most appropriate reagent based on their specific experimental design, target species, and detection method.

How do I determine the optimal dilution for my TARDBP antibody application?

Determining the optimal antibody dilution requires methodical testing and optimization for each specific application and sample type. For Western blotting applications, start with a range of dilutions (typically between 0.4-5 μg/mL as seen in validated protocols) using positive control samples like A431, HeLa, or RAW 264.7 cell lysates that are known to express TARDBP . For immunohistochemistry on paraffin-embedded tissue sections, a concentration range of 1.7-3 μg/mL has been validated for brain tissue samples . For immunofluorescence applications, start with 1-3 μg/mL as a working dilution . When optimizing, prepare a dilution series spanning at least three different concentrations (e.g., 1:500, 1:1000, 1:2000) and evaluate signal-to-noise ratio. The optimal dilution will provide strong specific staining of the target (generally nuclear localization for TARDBP in normal cells) with minimal background. Additional optimization may be necessary when changing experimental conditions, tissue types, or detection systems. Document all optimization steps and validated dilutions for reproducibility in future experiments.

What are the validated applications for TARDBP antibodies?

TARDBP antibodies have been validated for multiple experimental applications across different research contexts. Western blotting (WB) has been validated using human cell lines (A431, HeLa, K562, HepG2), mouse cell lines (RAW 264.7), and rat cell lines (NR8383) with specific bands detected at approximately 43-45 kDa under reducing conditions . Immunoprecipitation (IP) has been validated using HAP1 human near-haploid cell lines, demonstrating the ability to pull down TARDBP from complex protein mixtures . Immunofluorescence (IF) applications have been validated in fixed A431 human epithelial carcinoma cells and mouse splenocytes, with specific nuclear staining patterns observed . Immunohistochemistry with paraffin-embedded sections (IHCP) has been validated on human brain tissue (hippocampus and cortex) showing specific nuclear localization . Additionally, enzyme-linked immunosorbent assay (ELISA) applications have been validated for detecting human TARDBP in direct ELISA formats . For automated protein analysis, Simple Western™ systems have also been validated for detecting TARDBP in cell lysates . These diverse applications make TARDBP antibodies versatile tools for various research methodologies.

How should I prepare samples for optimal TARDBP detection by Western blot?

Optimal sample preparation for TARDBP Western blot analysis requires careful attention to protein preservation and extraction techniques. Begin by lysing cells or tissues in a buffer containing protease inhibitors to prevent protein degradation, which is particularly important for TARDBP due to its susceptibility to proteolytic cleavage. A recommended lysis buffer composition includes 50 mM Tris-HCl (pH 7.4), 150 mM NaCl, 1% NP-40 or Triton X-100, 0.5% sodium deoxycholate, 0.1% SDS, 1 mM EDTA, and a complete protease inhibitor cocktail. When working with tissues where TARDBP aggregates may be present (especially brain tissue from neurodegenerative disease models), consider sequential extraction methods using buffers of increasing solubilization strength to separate different protein fractions. For reducing conditions, add DTT (dithiothreitol) or β-mercaptoethanol to your sample buffer and heat samples at 95°C for 5 minutes before loading . Load 20-30 μg of total protein per lane for cell lines like A431, HeLa, or RAW 264.7 which express detectable levels of TARDBP. Transfer proteins to PVDF membranes (rather than nitrocellulose) for better protein retention and use Immunoblot Buffer Group 1 or 3 depending on your cell type as validated in published protocols . For detection, a specific band should be observed at approximately 43-45 kDa representing full-length TARDBP.

What controls should be included when using TARDBP antibodies in immunohistochemistry?

Robust control samples are essential for validating TARDBP antibody specificity in immunohistochemistry experiments. Include positive tissue controls known to express TARDBP such as human brain (cortex or hippocampus), breast, fallopian tube, testis, or skin samples . For disease-specific research, including tissue from patients with confirmed TDP-43 proteinopathies alongside age-matched controls can provide valuable comparison points. Negative controls should include tissues known to lack TARDBP expression or primary antibody omission tests on serial sections of your experimental tissue. Additionally, peptide competition assays, where the antibody is pre-incubated with purified TARDBP protein before application to tissue, can confirm binding specificity. For precise validation, include blocking peptide controls where available. When studying diseases with altered TARDBP localization (such as ALS or FTLD), include sections known to contain cytoplasmic TDP-43 aggregates alongside normal tissues with predominantly nuclear staining. This contrast will help validate the antibody's ability to detect pathological forms of the protein. Finally, consider dual-labeling with antibodies against different epitopes of TARDBP to further confirm specificity and potentially reveal differential detection of truncated or modified forms of the protein.

How can I distinguish between specific and non-specific bands in TARDBP Western blots?

Distinguishing between specific and non-specific bands in TARDBP Western blots requires systematic validation approaches. First, compare your observed band pattern with published literature; full-length TARDBP typically appears at approximately 43-45 kDa, though post-translational modifications or truncated forms may result in additional specific bands . Include positive control lysates from cell lines with confirmed TARDBP expression such as A431, HeLa, HepG2, K562 (human), RAW 264.7 (mouse), or NR8383 (rat) cell lines . Also consider running knockdown/knockout controls where TARDBP expression has been reduced or eliminated through siRNA or CRISPR/Cas9 methods—specific bands should show corresponding reduction in intensity. Peptide competition assays, where the antibody is pre-incubated with purified TARDBP protein before membrane incubation, can also help identify specific bands, which should diminish or disappear. For suspected cross-reactivity, use alternative antibodies targeting different TARDBP epitopes to confirm band patterns. When troubleshooting non-specific bands, optimize blocking conditions (consider switching between BSA and non-fat dry milk at 3-5%) and increase washing stringency. Adjusting antibody concentration and incubation time can also improve specificity. If working with brain tissue or disease models, be aware that C-terminal fragments (25-35 kDa) represent disease-specific cleavage products rather than non-specific bands .

What factors contribute to inconsistent TARDBP antibody performance across experiments?

Inconsistent TARDBP antibody performance can be attributed to several methodological and biological factors that require systematic troubleshooting. Antibody storage conditions significantly impact performance—repeated freeze-thaw cycles should be avoided by aliquoting the antibody upon first use, and storage temperature recommendations should be strictly followed. Different antibody lots may contain variations in activity and specificity, so maintaining detailed records of lot numbers and performing bridging experiments when transitioning between lots is advisable. Sample preparation variables also affect consistency; protein degradation during extraction, inconsistent fixation protocols, or variations in antigen retrieval methods can all alter epitope accessibility. For TARDBP specifically, its post-translational modifications (phosphorylation, ubiquitination, acetylation) may vary across experimental conditions and affect antibody recognition . When working with tissue samples, pre-analytical variables such as post-mortem interval and fixation duration critically impact immunoreactivity. Different detection systems (HRP-conjugated secondaries vs. fluorescent secondaries) have varying sensitivity thresholds and dynamic ranges. To enhance consistency, standardize all protocols including sample collection, processing times, buffer compositions, and incubation conditions. Incorporate internal controls in each experiment and consider using automated staining platforms for immunohistochemistry applications. Quantitative techniques like Western blotting should include loading controls and standard curves when possible.

How can TARDBP antibodies be used to study disease-specific protein modifications?

TARDBP antibodies can be strategically employed to investigate disease-specific modifications through complementary approaches targeting different protein states. For studying phosphorylation states, which are particularly relevant in ALS and FTLD where TARDBP is hyperphosphorylated, use phospho-specific antibodies alongside total TARDBP antibodies to calculate phosphorylation ratios . Immunoprecipitation with general TARDBP antibodies followed by immunoblotting with modification-specific antibodies (against phosphorylation, ubiquitination, acetylation, or SUMOylation sites) can reveal the proportion of modified protein in different experimental conditions. For detecting truncated forms of TARDBP common in pathological inclusions, combine antibodies targeting different epitopes (N-terminal vs. C-terminal) to identify specific fragmentation patterns. Sequential extraction protocols with buffers of increasing solubilization strength (e.g., low-salt, Triton X-100, sarkosyl, and urea fractions) followed by immunoblotting can differentiate soluble from aggregated TARDBP species. For microscopy applications, co-labeling with antibodies against TARDBP and specific modifications or aggregation markers (ubiquitin, p62, TIA-1) can reveal spatial relationships between protein states. Additionally, proximity ligation assays using TARDBP antibodies paired with modification-specific antibodies enable in situ visualization of modified protein with single-molecule sensitivity. When investigating novel modifications, mass spectrometry analysis of immunoprecipitated TARDBP can provide unbiased identification of post-translational modifications and their disease-associated changes.

What methodological approaches enable the study of TARDBP aggregation in disease models?

Studying TARDBP aggregation in disease models requires sophisticated methodological approaches spanning biochemical, microscopic, and functional analyses. Sequential extraction protocols represent a foundational biochemical approach, where tissues or cells are subjected to buffers with increasing solubilization strength (e.g., RIPA-soluble fraction followed by urea-soluble fraction) to separate normally soluble TARDBP from pathological aggregates. Western blotting of these fractions with TARDBP antibodies reveals distribution patterns between soluble and insoluble compartments . For microscopy-based approaches, immunohistochemistry using antibodies like clone BSB-166 can visualize changes in TARDBP subcellular localization, with nuclear clearing and cytoplasmic aggregation being hallmarks of pathology . Super-resolution microscopy techniques (STED, STORM) combined with appropriately conjugated TARDBP antibodies can reveal aggregate ultrastructure below the diffraction limit. Fluorescence recovery after photobleaching (FRAP) experiments using live-cell imaging with fluorescently tagged TARDBP can measure protein mobility changes associated with early aggregation. Filter trap assays, where cell lysates are passed through cellulose acetate membranes that trap large protein aggregates, followed by immunodetection with TARDBP antibodies, provide a quantitative measure of aggregation. For biochemical characterization, density gradient centrifugation of tissue homogenates followed by dot blotting with TARDBP antibodies can separate different sized aggregates. Additionally, conformation-specific antibodies that preferentially recognize misfolded TARDBP can be used alongside total TARDBP antibodies to distinguish pathological species from normal protein.

How can TARDBP antibodies be applied in multi-omics research approaches?

TARDBP antibodies can serve as powerful tools in multi-omics research by enabling the integration of protein-level data with transcriptomic, genomic, and metabolomic analyses. For proteomics applications, immunoprecipitation using validated TARDBP antibodies coupled with mass spectrometry (IP-MS) can identify the changing interactome of TARDBP under different conditions or disease states. Specifically, protein complexes can be isolated using antibodies like the mouse monoclonal E-10 (sc-376311) that has been validated for immunoprecipitation applications . In transcriptomic studies, ChIP-seq (Chromatin Immunoprecipitation followed by sequencing) using TARDBP antibodies can map genome-wide DNA binding sites, while CLIP-seq (Cross-Linking Immunoprecipitation) can identify RNA binding targets of TARDBP, providing insight into its regulatory networks. For spatial multi-omics, multiplexed immunofluorescence combining TARDBP antibodies with markers of cellular stress, RNA metabolism, or neurodegeneration can correlate protein localization with cell state or pathology. Proximity-based biotinylation approaches (BioID, APEX) using TARDBP fusion proteins followed by streptavidin pulldown and proteomic analysis can map the spatial proteome surrounding TARDBP under different conditions. For metabolomics integration, correlative microscopy approaches combining TARDBP immunostaining with mass spectrometry imaging can reveal relationships between TARDBP aggregation and metabolic alterations in tissues. When analyzing single-cell multi-omics data, TARDBP antibodies can be used in CyTOF or CODEX platforms to correlate protein expression with transcriptomic clusters. For all these applications, antibody validation is crucial—confirming specificity using multiple antibodies targeting different epitopes enhances confidence in multi-omics findings.

How do TARDBP antibodies perform in detecting pathological inclusions in ALS and FTLD tissues?

TARDBP antibodies exhibit variable performance in detecting pathological inclusions in ALS and FTLD tissues, with success depending on epitope specificity, tissue preparation, and detection methods. In these neurodegenerative conditions, TARDBP undergoes post-translational modifications including hyperphosphorylation, ubiquitination, and C-terminal fragmentation before forming cytoplasmic inclusions . For optimal detection of these pathological structures, antibodies recognizing the C-terminal region of TARDBP are often more effective as this region is contained in the disease-associated fragments. Monoclonal antibodies like clone BSB-166 have been validated for detecting both nuclear and cytoplasmic localization in disease tissues . Tissue preparation significantly impacts detection sensitivity—short fixation times (24-48 hours) in 10% neutral buffered formalin followed by standard paraffin embedding typically yield optimal results. Antigen retrieval methods are crucial, with heat-induced epitope retrieval in citrate buffer (pH 6.0) often providing superior unmasking of TARDBP epitopes in inclusion bodies. For visualization, amplification systems such as polymer-based detection methods or tyramide signal amplification can enhance sensitivity for detecting smaller or less abundant inclusions. Fluorescent approaches allow co-localization studies with other pathological markers such as ubiquitin or p62. When quantifying pathology, consider both the number of inclusions and the degree of nuclear clearing, as the latter represents the depletion of normal nuclear TARDBP that contributes to disease pathogenesis through loss-of-function mechanisms.

What considerations are important when using TARDBP antibodies in cancer research?

When utilizing TARDBP antibodies in cancer research, several specific considerations must be addressed to generate reliable and interpretable data. TARDBP has been identified as having context-dependent roles in cancer biology, functioning as both an oncogene in some cancers (melanoma, glioblastoma) and a tumor suppressor in others (e.g., through miRNA miR-500a-3p in rhabdomyosarcomas) . Therefore, antibody selection should be guided by the specific cancer type under investigation. For studying TARDBP's role in cancer metabolism, antibodies detecting the full-length protein are generally appropriate as TARDBP regulates RNA associated with glucose and lipid metabolism . When investigating subcellular localization changes in cancer cells, immunofluorescence using antibodies validated for both nuclear and cytoplasmic detection is recommended, as TARDBP's function can vary based on its cellular compartmentalization. For quantitative assessments in cancer tissues, immunohistochemistry using antibodies like BSB-166 has been validated on diverse cancer types including lung adenocarcinoma . When studying TARDBP's interaction with miRNAs (such as miR-152, miR-500a-3p) in cancer cells, combining RNA-protein co-immunoprecipitation with TARDBP antibodies followed by qRT-PCR for specific miRNAs can reveal functional interactions. For high-throughput analyses, tissue microarray staining with carefully validated TARDBP antibodies can assess expression across multiple patient samples simultaneously. Cancer tissue heterogeneity necessitates analyzing multiple regions within tumors, and comparing TARDBP expression between tumor core, invasive front, and adjacent normal tissue can provide insights into its role in cancer progression.

How can TARDBP antibodies be used to investigate the protein's role in cellular stress responses?

TARDBP antibodies provide powerful tools for investigating the protein's dynamic behavior during cellular stress responses through multiple experimental approaches. Under stress conditions, TARDBP can relocalize from the nucleus to the cytoplasm and participate in stress granule formation—a process implicated in both adaptive stress responses and pathological aggregation in neurodegenerative diseases . To track this stress-induced translocation, live-cell imaging using fluorescently-tagged antibody fragments (Fabs) against TARDBP can monitor real-time localization changes in response to stressors such as oxidative stress, heat shock, or ER stress. For fixed-cell analyses, co-immunofluorescence using TARDBP antibodies alongside stress granule markers (G3BP1, TIA-1, eIF3) after stress induction reveals recruitment kinetics and composition of these membrane-less organelles. Biochemical fractionation of stressed cells into nuclear and cytoplasmic compartments followed by Western blotting with TARDBP antibodies provides quantitative assessment of translocation efficiency. To investigate posttranslational modifications associated with stress responses, immunoprecipitation with general TARDBP antibodies followed by modification-specific antibody detection (phospho-specific, ubiquitin-specific) can reveal stress-induced changes in protein state. For studying TARDBP's stress-related RNA targets, CLIP (Cross-Linking Immunoprecipitation) using validated TARDBP antibodies before and after stress exposure identifies dynamic changes in bound transcripts. When investigating the relationship between stress responses and disease pathology, proximity ligation assays combining TARDBP antibodies with antibodies against stress response proteins can visualize and quantify specific interactions that may be altered in disease states.

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.