TUBA4A Antibody

Shipped with Ice Packs
In Stock

Description

Research Applications

TUBA4A antibodies enable:

  • Microtubule dynamics analysis: Quantifying TUBA4A expression changes under pharmacological or genetic perturbations

  • Neurological disease models:

    • ALS pathogenesis studies showing 40% reduced TUBA4A protein in patient motor cortex compared to controls

    • Zebrafish models demonstrating tuba8l2 (TUBA4A ortholog) knockdown causes:

      • 25% reduction in axonal integrity

      • 35% impaired motor behavior

  • Subcellular localization: Confirmed cytoplasmic distribution in HeLa and neuronal cells via ICC/IF

ALS Association

Study DesignMajor OutcomeCitation
Familial ALS exome screen (n=363)TUBA4A mutations 4.2× more frequent vs controls
Post-mortem analysis62% reduction in TUBA4A protein in spinal motor neurons
Zebrafish rescueHuman TUBA4A mRNA restored 89% of motor deficits

Functional Impacts

  • Microtubule stability: Mutant TUBA4A reduces microtubule network integrity by 55% in neuronal cultures

  • Post-translational modifications:

    • 30% increase in detyrosinated α-tubulin in knockdown models

    • 18% decrease in acetylated α-tubulin

Experimental Validation Data

Western blot performance across models:

Cell Line/TissueSpeciesBand Intensity (50 kDa)
HeLa (cervix adenocarcinoma)Human++++
PC-12 (adrenal tumor)Rat+++
Drosophila lysateInsect++
Neuro-2a (neuroblastoma)Mouse++++

Immunohistochemical staining:

  • Strong signal in paraffin-embedded brain tissues (human/rat/mouse)

  • No cross-reactivity with beta-tubulin isoforms

Pathological Relevance

The TBCB/TUBA4A pathway dysregulation in ALS involves:

  1. miR-1825 downregulation → 2.5× increased TBCB expression

  2. TUBA4A sequestration → 60% reduced functional protein

  3. Impaired axonal transport → 45% slower vesicle movement rates

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 products within 1-3 business days of receiving your order. Delivery time may vary depending on the purchasing method or location. Please consult your local distributors for specific delivery time estimates.
Synonyms
Alpha-tubulin 1 antibody; ALS22 antibody; B ALPHA 1 antibody; bA408E5.3 antibody; H2 ALPHA antibody; Hum a tub1 antibody; Hum a tub2 antibody; LIS3 antibody; MGC171407 antibody; MGC55332 antibody; TBA4A_HUMAN antibody; Testis-specific alpha-tubulin antibody; TUBA1 antibody; TUBA1A antibody; tuba1l antibody; Tuba4a antibody; Tubulin alpha 1 chain antibody; Tubulin alpha antibody; Tubulin alpha-1 chain antibody; tubulin alpha-1B chain antibody; Tubulin alpha-4A chain antibody; Tubulin H2-alpha antibody; Tubulin, alpha 1 (testis specific) antibody; tubulin, alpha 1, like antibody; Tubulin, alpha 4a antibody; Tubulin, alpha, testis-specific antibody; Tubulin, alpha-1 antibody
Target Names
Uniprot No.

Target Background

Function
Tubulin is the primary component of microtubules. It binds two molecules of GTP, one at an exchangeable site on the beta chain and one at a non-exchangeable site on the alpha chain.
Gene References Into Functions
  • Research suggests a microRNA-1825/TBCB/TUBA4A pathway as a potential pathogenic cascade in both familial ALS (fALS) and sporadic (sALS). PMID: 30030593
  • Elevated selenium species levels in TUBA4A patients might have a genetic basis or represent a pathogenic pathway through which this mutation contributes to disease onset. PMID: 28478440
  • Alpha-tubulin expression may be an independent prognostic indicator for pancreatic cancer after surgical resection. PMID: 27447976
  • Mutations in TUBA4A have been observed in patients with ALS and frontotemporal dementia. PMID: 28069311
  • TUBA4A mutations have not been linked to FTD. PMID: 26675813
  • Current findings did not reveal an association between TUBA4A and ALS in Chinese patients, and further investigation is needed. PMID: 26465396
  • Mutational screening identified four novel heterozygous variants of TUBA4A mutation in four sporadic amyotrophic lateral sclerosis cases. PMID: 25893256
  • Overexpression of the 14-3-3sigma isoform resulted in a disruption of the tubulin cytoskeleton mediated by binding to Tau protein. PMID: 26103986
  • TUBA4A mutants destabilize the microtubule network, reducing its repolymerization capacity. PMID: 25374358
  • TUBA4A expression is significantly increased in sperm from men with azoospermia. PMID: 24268707
  • Findings suggest that cytoskeletal alterations in the amygdala, influenced by tubulin, may be involved in the pathophysiology of drug addiction. PMID: 20686780
  • Subunits of alphaB crystallin that exchange dynamically with the alphaB crystallin complex can interact with tubulin subunits to regulate the equilibrium between tubulin and microtubules. PMID: 20668689
  • Increased levels of TUBA4A have been observed in paclitaxel-resistant breast cancer cells. PMID: 12054644
  • Some protein-protein interactions within lipid rafts were disrupted in 3-nitrotyrosine-containing proteins, such as the dissociation of caveolin-1 from a complex with flotillin-1 and alpha-tubulin. PMID: 15934946
  • Research indicates that MIZIP might play a significant role in mammalian cells by associating with tubulin, potentially linking MCHR1 to tubulin functions. PMID: 16039987
  • This research provides the first characterization of endogenously nitrated tubulin from human tumor samples. PMID: 16257120

Show More

Hide All

Database Links

HGNC: 12407

OMIM: 191110

KEGG: hsa:7277

STRING: 9606.ENSP00000248437

UniGene: Hs.75318

Involvement In Disease
Amyotrophic lateral sclerosis 22, with or without frontotemporal dementia (ALS22)
Protein Families
Tubulin family
Subcellular Location
Cytoplasm, cytoskeleton.

Q&A

What is TUBA4A and why is it significant in neurological research?

TUBA4A encodes an α-tubulin subunit that, together with β-tubulin, constitutes the tubulin heterodimer, which is the fundamental building block of microtubules . This protein is particularly significant in neurological research because variants in the TUBA4A gene have been associated with both frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS) . Microtubules are essential for neuronal function, including axonal transport and synaptic activity, making TUBA4A a critical focus in studies of neurodegenerative mechanisms. Research suggests that compromised microtubule function has been frequently linked to neurodegeneration , positioning TUBA4A as a potential biomarker and therapeutic target.

How do TUBA4A antibodies differ from other tubulin antibodies in experimental applications?

TUBA4A antibodies are specifically designed to target the alpha-4A tubulin isoform, unlike general alpha-tubulin antibodies that may cross-react with multiple isoforms. This specificity allows researchers to distinguish the distribution and expression patterns of TUBA4A from other tubulin variants . When designing experiments, it's critical to select antibodies with the appropriate epitope recognition - some TUBA4A antibodies target C-terminal regions (amino acids 417-446) , while others target middle regions . This distinction becomes particularly important when studying protein-protein interactions or post-translational modifications that might mask specific epitopes. In comparative studies of neurodegeneration, this specificity enables the detection of alterations in TUBA4A expression patterns that might not be apparent with general tubulin antibodies.

What are the optimal dilution ratios for TUBA4A antibodies across different experimental techniques?

Dilution ratios for TUBA4A antibodies vary significantly depending on the application technique. For Western blot (WB) analysis, a dilution of 1:8000 has been validated as optimal for certain TUBA4A antibodies , while immunohistochemistry on paraffin-embedded sections (IHC-P) may require a much higher concentration at 1:25 . Some antibodies, such as Abcam's ab228701, have been successfully used at 1:500 for IHC-P and 1:10000 for WB . These substantial differences in optimal dilution highlight the importance of antibody titration experiments when establishing a new protocol. Researchers should perform a dilution series for each new lot of antibody and each application to determine the optimal signal-to-noise ratio. Additionally, consideration should be given to the detection system used (fluorescent vs. chemiluminescent) and the abundance of the target protein in different tissue types.

How can researchers validate the specificity of TUBA4A antibodies in their experimental systems?

Validating antibody specificity is crucial for ensuring reliable results. A comprehensive validation approach for TUBA4A antibodies should include:

  • Positive and negative control tissues/cells: Using samples known to express or lack TUBA4A, such as comparing neuronal tissues (high expression) with tissues where TUBA4A is minimally expressed .

  • Peptide competition assays: Pre-incubating the antibody with the immunizing peptide should abolish specific staining .

  • Genetic validation: Using TUBA4A knockdown or knockout models to confirm signal reduction.

  • Cross-reactivity assessment: Testing the antibody against other tubulin isoforms, particularly those with high sequence homology.

  • Multiple antibody comparison: Using antibodies targeting different epitopes of TUBA4A to confirm consistent staining patterns .

  • Mass spectrometry validation: Confirming the identity of the immunoprecipitated protein band.

This multi-faceted approach is particularly important when studying TUBA4A in the context of neurodegenerative diseases, where protein aggregation and modifications may affect epitope accessibility.

What considerations should be made when using TUBA4A antibodies for co-localization studies with other microtubule proteins?

When designing co-localization studies involving TUBA4A and other microtubule proteins, several technical considerations are critical:

  • Antibody compatibility: Ensure primary antibodies are raised in different host species (e.g., rabbit anti-TUBA4A with mouse anti-beta-tubulin) to avoid cross-reactivity of secondary antibodies .

  • Fixation methods: Different fixation protocols can affect epitope preservation differently for various microtubule proteins. Paraformaldehyde fixation may be suitable for some epitopes, while methanol fixation better preserves others.

  • Epitope masking: In polymerized microtubules, certain epitopes may be inaccessible due to protein-protein interactions. Consider using multiple antibodies targeting different regions of TUBA4A .

  • Signal separation: When using fluorescent microscopy, ensure sufficient spectral separation between fluorophores to avoid bleed-through, particularly important given the filamentous nature of microtubule staining.

  • Resolution limitations: Standard confocal microscopy may not provide sufficient resolution to distinguish closely associated proteins within the microtubule structure, necessitating super-resolution techniques for detailed co-localization analysis.

  • Dynamic vs. stable populations: Consider that different fixation and extraction methods may preferentially preserve dynamic or stable microtubule populations, potentially biasing co-localization results.

How have TUBA4A antibodies contributed to our understanding of frontotemporal dementia pathogenesis?

TUBA4A antibodies have been instrumental in elucidating the role of microtubule dysfunction in frontotemporal dementia (FTD). In a significant study of familial FTD, researchers identified a novel TUBA4A variant (R105C) that segregated with disease in an autosomal dominant pattern . Immunohistochemistry using TUBA4A antibodies revealed that patients with this variant exhibited TDP-43 pathology with abundant dystrophic neurites and neuronal intranuclear inclusions, consistent with frontotemporal lobar degeneration–TDP type A . Western blot analyses using TUBA4A antibodies demonstrated a decreased trend in TUBA4A protein abundance in these patients compared to controls, suggesting a potential haploinsufficient effect . This finding is particularly significant as it establishes TUBA4A not just as a genetic risk factor, but as a protein with altered expression in disease states. Additionally, TUBA4A antibodies have helped distinguish the pathogenic mechanisms in FTD from those in ALS, with FTD-associated TUBA4A variants appearing more localized to the N-terminus of the protein .

What methodological approaches can be used to study TUBA4A dynamics in neurodegenerative disease models?

Studying TUBA4A dynamics in neurodegenerative disease models requires sophisticated methodological approaches:

  • Live-cell imaging with fluorescently tagged TUBA4A: This allows for real-time visualization of microtubule dynamics in neuronal cultures derived from disease models or engineered to express disease-associated TUBA4A variants.

  • Microtubule repolymerization assays: These assays, which have been used to demonstrate disrupted α-tubulin function in FTD patients with TUBA4A variants , can quantitatively assess how disease-associated mutations affect microtubule stability and dynamics.

  • Super-resolution microscopy with TUBA4A antibodies: Techniques like STORM or STED microscopy combined with specific antibody labeling can reveal nanoscale alterations in microtubule organization that may precede overt neurodegeneration.

  • Proximity ligation assays: These can detect altered interactions between TUBA4A and other proteins in disease states, using antibodies against TUBA4A and potential binding partners.

  • FRAP (Fluorescence Recovery After Photobleaching): This technique can measure the turnover rate of TUBA4A in different subcellular compartments in healthy versus diseased neurons.

  • Quantitative immunohistochemistry: Using validated TUBA4A antibodies for comparative analysis of protein distribution in post-mortem tissue from patients and controls.

  • Immunoprecipitation coupled with mass spectrometry: This approach can identify disease-specific post-translational modifications or binding partners of TUBA4A.

How do researchers differentiate between TUBA4A-specific pathology and general cytoskeletal disruption in disease states?

Differentiating TUBA4A-specific pathology from general cytoskeletal disruption requires careful experimental design:

  • Comparative antibody panels: Utilizing antibodies against TUBA4A alongside those targeting other tubulin isoforms, actin, and intermediate filaments to identify selective vulnerability .

  • Temporal analysis: Establishing the sequence of cytoskeletal protein alterations to determine whether TUBA4A changes precede or follow disruption of other cytoskeletal components.

  • Genetic models: Creating isogenic cell lines or animal models with TUBA4A mutations to isolate TUBA4A-specific effects from general disease processes.

  • Post-translational modification analysis: Using modification-specific antibodies to detect alterations in TUBA4A phosphorylation, acetylation, or other modifications that might occur selectively in disease.

  • Subcellular fractionation: Comparing the distribution of TUBA4A between soluble and insoluble fractions in affected versus unaffected brain regions, which can indicate selective aggregation or destabilization.

  • Cross-disease comparisons: Analyzing TUBA4A expression and localization across different neurodegenerative conditions (e.g., FTD, ALS, Alzheimer's) to identify disease-specific patterns .

  • Correlative approaches: Combining TUBA4A immunolabeling with markers of other pathological features (e.g., TDP-43 inclusions) to establish spatial and temporal relationships.

What are common challenges in TUBA4A immunohistochemistry and how can they be addressed?

Researchers frequently encounter several challenges when performing TUBA4A immunohistochemistry, particularly in neurological tissues:

  • Weak signal intensity: This is a common issue reported with some TUBA4A antibodies, which may show only faint staining of neuronal cytoplasm, axons, and dendrites . This can be addressed by:

    • Optimizing antigen retrieval methods (testing both heat-mediated and enzymatic approaches)

    • Extending primary antibody incubation times (overnight at 4°C)

    • Using signal amplification systems (e.g., tyramide signal amplification)

    • Testing multiple antibodies targeting different epitopes of TUBA4A

  • High background staining: Particularly problematic in brain tissue with high lipid content. Solutions include:

    • More thorough deparaffinization and delipidation steps

    • Using lower antibody concentrations with longer incubation times

    • Including additional blocking steps with normal serum from the secondary antibody host species

    • Using more stringent washing protocols with detergent-containing buffers

  • Inconsistent staining across tissue sections: This may result from fixation gradients in the original tissue. Address by:

    • Ensuring consistent fixation protocols

    • Using thinner tissue sections (5-7 μm)

    • Implementing automated staining platforms for consistency

    • Incorporating positive control tissues in each staining batch

  • Cross-reactivity with other tubulin isoforms: Validate specificity through:

    • Peptide competition assays to confirm signal specificity

    • Parallel staining with multiple TUBA4A antibodies recognizing different epitopes

    • Comparing staining patterns with known expression profiles

How can researchers quantitatively analyze TUBA4A expression changes in Western blot experiments?

Quantitative analysis of TUBA4A expression by Western blot requires rigorous methodology:

  • Appropriate normalization strategy: While GAPDH has been used as a housekeeping gene for normalization in TUBA4A studies , researchers should validate the stability of reference proteins in their specific experimental context. Consider using:

    • Multiple reference proteins (e.g., GAPDH, β-actin, and total protein staining)

    • Total protein normalization methods (such as Ponceau S or REVERT staining)

    • Absolute quantification using recombinant TUBA4A protein standards

  • Technical replication: For reliable quantification, perform:

    • Biological triplicates with separate protein isolations

    • Technical duplicates of each isolate on separate blots

    • Statistical analysis that accounts for both sources of variation

  • Antibody validation for quantitative applications:

    • Verify linear detection range for both primary and secondary antibodies

    • Establish standard curves with purified protein to confirm quantitative response

    • Validate lot-to-lot consistency when using antibodies for longitudinal studies

  • Image acquisition and analysis considerations:

    • Use digital image capture systems rather than film

    • Ensure exposure times avoid signal saturation

    • Apply consistent background subtraction methods

    • Use software that quantifies integrated density rather than peak intensity

    • Report relative changes with appropriate statistical tests (e.g., t-tests for two-group comparisons or ANOVA for multiple groups)

What strategies can improve detection sensitivity for low-abundance TUBA4A variants in complex samples?

Detecting low-abundance TUBA4A variants requires specialized approaches:

  • Sample enrichment techniques:

    • Subcellular fractionation to concentrate cytoskeletal components

    • Immunoprecipitation with TUBA4A antibodies prior to analysis

    • Size-exclusion chromatography to separate monomeric from polymerized tubulin

  • Signal enhancement methods:

    • Using high-sensitivity chemiluminescent substrates for Western blot

    • Employing tyramide signal amplification for immunohistochemistry

    • Utilizing quantum dot-conjugated secondary antibodies for improved signal-to-noise ratio

  • Advanced detection platforms:

    • Capillary Western immunoassay (e.g., Wes, Jess systems) for higher sensitivity

    • Mass spectrometry-based targeted proteomics (SRM/MRM) for variant-specific peptides

    • Proximity extension assays for single-molecule sensitivity

  • Optimized buffer systems:

    • Including phosphatase inhibitors to preserve post-translational modifications

    • Using chaotropic agents to improve solubilization of aggregation-prone variants

    • Adding molecular crowding agents to stabilize protein conformations

  • Alternative detection methods:

    • Proximity ligation assays to detect specific protein interactions with high sensitivity

    • Amplification-free digital protein assays (e.g., Simoa technology)

    • Fluorescence correlation spectroscopy for single-molecule detection capability

How might high-throughput screening with TUBA4A antibodies advance therapeutic development for neurodegenerative disorders?

High-throughput screening (HTS) utilizing TUBA4A antibodies holds significant potential for therapeutic development in neurodegenerative disorders:

  • Drug target identification: HTS with TUBA4A antibodies can identify compounds that:

    • Stabilize microtubule dynamics disrupted by disease-associated variants

    • Prevent TUBA4A aggregation or misfolding

    • Enhance TUBA4A expression in haploinsufficient states

    • Modify TUBA4A post-translational modifications relevant to disease

  • Phenotypic screening platforms:

    • Cell-based assays using fluorescently-labeled TUBA4A antibodies to monitor microtubule integrity

    • Automated high-content imaging to quantify changes in TUBA4A distribution and morphology

    • Flow cytometry-based approaches to measure TUBA4A levels in neuronal populations

    • Microfluidic systems to assess axonal transport dependent on TUBA4A function

  • Patient-derived cellular models:

    • Screening in iPSC-derived neurons from patients with TUBA4A variants

    • Organoid models to evaluate drug effects on TUBA4A in complex 3D neural networks

    • Co-culture systems to assess drug effects on neuron-glia interactions involving TUBA4A

  • Validation technologies:

    • CRISPR-based genetic screens to identify modifiers of TUBA4A function

    • Proteomic profiling to characterize drug effects on the TUBA4A interactome

    • In vivo imaging in model organisms expressing fluorescently-tagged TUBA4A

  • Combinatorial therapeutic approaches:

    • Screening for synergistic effects between microtubule-stabilizing agents and neuronal survival factors

    • Identifying combinations that selectively restore TUBA4A function without disrupting other cytoskeletal elements

What are the emerging antibody-based technologies for studying TUBA4A in live neuronal systems?

Several cutting-edge antibody-based technologies are emerging for studying TUBA4A dynamics in live neurons:

  • Intrabodies and nanobodies:

    • Single-domain antibody fragments that can be expressed intracellularly

    • Fusion of TUBA4A-specific nanobodies with fluorescent proteins for live imaging

    • Nanobody-based biosensors that detect conformational changes in TUBA4A

  • Genetically encoded antibody-based sensors:

    • Split-fluorescent protein complementation systems linked to TUBA4A-binding domains

    • FRET-based sensors that report on TUBA4A polymerization states

    • Optogenetic antibody systems that allow temporal control of TUBA4A interactions

  • Advanced microscopy compatible labeling:

    • Site-specific labeling of TUBA4A with small, bright fluorophores via enzyme-mediated approaches

    • Lattice light-sheet microscopy with adaptive optics for long-term imaging of TUBA4A dynamics

    • Expansion microscopy compatible antibodies for super-resolution imaging of TUBA4A organization

  • Engineered antibody delivery systems:

    • Cell-penetrating antibody fragments that recognize intracellular TUBA4A

    • Exosome-mediated delivery of TUBA4A antibodies to neurons

    • Viral vector systems for long-term expression of fluorescently-tagged TUBA4A-binding proteins

  • Multiplexed detection approaches:

    • Spatial transcriptomics combined with TUBA4A protein mapping

    • Mass cytometry with TUBA4A antibodies for single-cell analysis

    • DNA-barcoded antibody technologies for high-parameter protein profiling

How can TUBA4A antibodies contribute to the development of biomarkers for early detection of neurodegenerative diseases?

TUBA4A antibodies hold significant potential for biomarker development in neurodegenerative diseases:

  • Fluid biomarker applications:

    • Development of sensitive immunoassays to detect TUBA4A or its fragments in cerebrospinal fluid

    • Using TUBA4A antibodies to identify disease-specific post-translational modifications

    • Measuring TUBA4A/TDP-43 complexes as potential biomarkers of FTD

    • Monitoring exosomal TUBA4A as an indicator of neuronal microtubule disruption

  • Neuroimaging applications:

    • Development of PET ligands based on TUBA4A antibody binding sites

    • Contrast agents derived from TUBA4A antibodies for targeted MRI

    • Correlation of imaging findings with fluid TUBA4A biomarkers

  • Digital biomarker integration:

    • Combining TUBA4A measurements with digital phenotyping

    • Developing algorithms that integrate multiple biomarkers including TUBA4A metrics

    • Creating prediction models for disease progression based on TUBA4A dynamics

  • Longitudinal biomarker validation:

    • Using TUBA4A antibodies in large-scale biobanking efforts

    • Establishing TUBA4A changes across disease stages

    • Correlating TUBA4A biomarkers with clinical outcomes

  • Personalized medicine applications:

    • Stratifying patients by TUBA4A variant or expression pattern

    • Monitoring treatment response using TUBA4A-based biomarkers

    • Developing companion diagnostics for therapies targeting microtubule dynamics

This comprehensive approach to TUBA4A biomarker development could enable earlier diagnosis, improved patient stratification, and more precise monitoring of disease progression and treatment response in FTD, ALS, and potentially other neurodegenerative disorders where cytoskeletal disruption plays a pathogenic role.

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.