BT1 Antibody

Shipped with Ice Packs
In Stock

Description

BT1-4F10: Bat Immunoglobulin Antibody

BT1-4F10 is a mouse-derived monoclonal antibody (mAb) designed to detect immunoglobulins in microchiropteran bats. Key characteristics include:

PropertyDetails
Host SpeciesMouse
TargetImmunoglobulin light chain (κ and λ) in bats
ReactivityMicrochiropteran bats (e.g., big brown, little brown, hoary bats)
ApplicationsELISA, Western blot, immunofluorescence, flow cytometry, immunohistochemistry
SpecificityRecognizes all bat immunoglobulin classes (IgG, IgM, IgA, IgD, IgE)
Research UseDetects rabies infection, studies bat immune responses to pathogens like Geomyces destructans (white-nose syndrome)

Developed by Health Research, Inc., BT1-4F10 enables rabies diagnosis in bats without viral neutralization assays, streamlining surveillance efforts . It also facilitates studies on bat immunity, a critical area given bats’ role as reservoirs for viruses like SARS-CoV and rabies .

BT1 (BCMA×CD3): Bispecific T-Cell Engager Antibody

BT1 (BCMA×CD3) is an IgG-like bispecific antibody targeting B-cell maturation antigen (BCMA) on tumor cells and CD3 on T cells. It is engineered for multiple myeloma and other BCMA-positive cancers.

PropertyDetails
TargetsBCMA (tumor cells) and CD3 (T cells)
MechanismRedirects T cells to lyse BCMA-expressing cancer cells
Clinical FindingsInduces T-cell activation and tumor cell death but triggers cytokine release syndrome (CRS)
Comparative EfficacyLess effective than NK-cell engager BK1 (BCMA×CD16) in reducing cytokine storms

BT1-4F10 in Bat Immunology

  • Identifies rabies-infected bats via immunoglobulin light chain detection in serum assays .

  • Enables species-specific studies on bat immune responses, critical for understanding zoonotic disease dynamics .

BT1 (BCMA×CD3) in Oncology

  • Demonstrates dose-dependent tumor cell killing in multiple myeloma models .

  • Combinatorial use with BK1 reduces proinflammatory cytokines (IFN-γ, TNF) by 40–60% compared to monotherapy .

Challenges and Future Directions

  • BT1-4F10: Limited to microchiropteran bats; no cross-reactivity with megabats or non-bat species .

  • BT1 (BCMA×CD3): Requires optimization to minimize CRS while retaining efficacy.

Product Specs

Buffer
Preservative: 0.03% Proclin 300; Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
14-16 weeks (Made-to-order)
Synonyms
BT1 antibody; At5g63160 antibody; MDC12.13BTB/POZ and TAZ domain-containing protein 1 antibody; BTB and TAZ domain protein 1 antibody
Target Names
BT1
Uniprot No.

Target Background

Function
This antibody targets a protein that may function as a substrate-specific adapter within a CUL3-RBX1-BTB E3 ubiquitin-protein ligase complex. This complex mediates the ubiquitination and subsequent proteasomal degradation of target proteins. The target protein itself is also a substrate for degradation via the 26S proteasome pathway. It may also play a role in gametophyte development.
Gene References Into Functions
PMID: 27208309, Members of the BTB gene family of scaffold proteins function as conserved negative regulators of nitrate uptake genes and nitrogen use efficiency in plants., .
Database Links

KEGG: ath:AT5G63160

STRING: 3702.AT5G63160.1

UniGene: At.27672

Subcellular Location
Nucleus. Cytoplasm.
Tissue Specificity
Preferentially expressed in young leaves, roots and stems.

Q&A

What is TBK1 and why is it important in research?

TBK1 (TANK-binding kinase 1) is a serine-threonine protein kinase that has emerged as a significant research target due to its connections to multiple diseases, including amyotrophic lateral sclerosis and frontotemporal dementia. As a key protein kinase, TBK1 plays crucial roles in various cellular pathways, making it an important subject for research investigations. The research community has faced challenges with TBK1 studies partly due to limitations in well-characterized antibodies for this protein. Reproducible research on TBK1 requires reliable antibodies for consistent and accurate detection and characterization of this protein in various experimental contexts .

The scientific significance of TBK1 extends beyond neurodegenerative diseases, as researchers continue to explore its roles in immunity and other cellular processes. The availability of specific and reliable antibodies against TBK1 is therefore essential for advancing our understanding of this protein's functions and potential therapeutic applications. Researchers should approach TBK1 studies with careful consideration of the antibodies used to ensure validity and reproducibility of their findings .

How can I validate the specificity of a TBK1 antibody for my research?

Antibody validation is critical for ensuring research reproducibility. The gold standard for validating TBK1 antibodies involves using isogenic knockout (KO) controls alongside wild-type (WT) cells in a comparative analysis. This approach allows researchers to definitively determine antibody specificity by confirming the absence of signal in KO cells. For comprehensive validation, researchers should test antibodies in multiple applications including immunoblot (Western blot), immunofluorescence, and immunoprecipitation to ensure specificity across different experimental contexts .

A particularly effective validation strategy involves the mosaic approach, where WT cells are labeled with one fluorescent dye (e.g., green) and KO cells with another (e.g., far-red), then plated together and visualized in the same field of view. This reduces imaging and analysis biases, providing more reliable validation results. Additionally, researchers should validate antibodies by checking for appropriate molecular weight bands in Western blots, correct cellular localization patterns in immunofluorescence, and specific pull-down in immunoprecipitation assays. Documentation of all validation steps enhances transparency and reproducibility in research .

What are the technical considerations for selecting an appropriate TBK1 antibody?

When selecting a TBK1 antibody, researchers should consider several critical factors beyond basic reactivity. First, determine the specific application needs (Western blot, immunofluorescence, immunoprecipitation) as antibodies often perform differently across applications. Check if the antibody has been validated using knockout controls specifically for your application of interest, as validation in one application doesn't guarantee performance in another. Consider the antibody type (monoclonal vs. polyclonal) based on your experimental requirements – monoclonals typically offer higher specificity while polyclonals may provide stronger signals but potentially more background .

The host species of the antibody is another important consideration, especially for multi-labeling experiments where avoiding cross-reactivity is essential. Review the epitope information to ensure it matches your experimental design, particularly if you're studying specific protein domains or post-translational modifications. Additionally, consider the antibody format (whole IgG, Fab fragments, etc.) based on your specific application needs. Finally, whenever possible, select antibodies with Research Resource Identifiers (RRIDs) to ensure proper citation and reproducibility in your research . Following these considerations will help ensure selection of the most appropriate TBK1 antibody for your specific research needs.

What are the optimal storage and handling conditions for TBK1 antibodies?

Proper storage and handling of TBK1 antibodies are essential for maintaining their specificity and activity. While specific recommendations may vary slightly between manufacturers, most antibodies perform best when stored at -20°C for long-term storage, with working aliquots kept at 4°C to minimize freeze-thaw cycles. Repeated freeze-thaw cycles can significantly reduce antibody activity and should be avoided by preparing single-use aliquots. Additionally, antibodies should be protected from light, especially those conjugated with fluorophores, to prevent photobleaching .

For handling during experiments, researchers should maintain antibodies at the appropriate temperature according to the manufacturer's recommendations. Dilution buffers should typically contain a stabilizing protein (such as BSA) and preservatives to prevent microbial contamination during storage. When preparing working solutions, centrifuge the antibody vial before opening to collect liquid at the bottom, and use sterile technique when handling to prevent contamination. For long experimental procedures, keeping antibodies on ice can help preserve their activity. Documentation of storage conditions, handling procedures, and lot numbers is important for experimental reproducibility and troubleshooting if unexpected results occur .

How do you optimize immunoprecipitation protocols for studying TBK1 protein interactions?

Optimizing immunoprecipitation (IP) protocols for TBK1 requires careful consideration of several parameters. First, select a TBK1 antibody specifically validated for IP applications, as not all antibodies that work for Western blot will perform well in IP. The choice between native and denaturing conditions is crucial - while native conditions preserve protein interactions, denaturing conditions may increase epitope accessibility. For studying TBK1 protein interactions, native conditions are generally preferred. The lysis buffer composition significantly impacts success, with RIPA or NP-40-based buffers commonly used for TBK1 IP, supplemented with protease and phosphatase inhibitors to preserve protein integrity .

The antibody-to-lysate ratio requires careful optimization, typically starting with 2-5 μg of antibody per 500-1000 μg of total protein. For TBK1 studies, magnetic beads often provide advantages over agarose beads due to reduced background and gentle handling. Incubation time and temperature also affect IP efficiency - overnight incubation at 4°C generally yields optimal results for TBK1 interactions. Washing steps must be stringent enough to remove non-specific interactions without disrupting specific ones, usually requiring 3-5 washes with decreasing salt concentrations. For detecting co-immunoprecipitated proteins, sensitive detection methods like mass spectrometry may reveal novel interacting partners, while Western blotting confirms specific interactions. Finally, including appropriate controls (such as IgG control and input samples) is essential for accurate interpretation of results .

What approaches are effective for studying TBK1 in the context of cellular signaling pathways?

Studying TBK1 in cellular signaling pathways requires multilevel experimental approaches. Phospho-specific antibodies are particularly valuable for monitoring TBK1 activation, as TBK1 undergoes autophosphorylation at Ser172 during activation. Western blotting with phospho-specific and total TBK1 antibodies can reveal activation dynamics under different stimulation conditions. Live-cell imaging using fluorescently tagged TBK1 combined with pathway-specific stimulation allows real-time visualization of TBK1 localization and trafficking. Additionally, proximity ligation assays (PLA) can detect endogenous protein-protein interactions between TBK1 and its binding partners with high specificity and sensitivity .

For examining TBK1's role in specific pathways, selective inhibitors combined with pathway readouts provide valuable insights. RNA interference (siRNA or shRNA) or CRISPR-Cas9-mediated knockdown/knockout of TBK1 followed by pathway analysis can establish causality in signaling events. Reconstitution experiments where TBK1 knockout cells are complemented with wild-type or mutant TBK1 can determine the functional significance of specific domains or phosphorylation sites. Mass spectrometry-based phosphoproteomics comparing wild-type and TBK1-deficient cells can identify downstream substrates. For comprehensive pathway mapping, combining multiple techniques such as co-immunoprecipitation, chromatin immunoprecipitation (ChIP), and transcriptome analysis provides a more complete picture of TBK1's role in specific signaling cascades .

How can TBK1 antibodies be used for distinguishing between different conformational states of the protein?

Distinguishing between different conformational states of TBK1 represents an advanced research challenge that requires specialized antibody approaches. Conformation-specific antibodies can be developed and selected to recognize distinct structural states of TBK1, such as its active versus inactive conformations. These antibodies typically target epitopes that are exposed or hidden depending on the protein's conformational state. For TBK1, which undergoes significant conformational changes upon activation, antibodies targeting the activation loop or regions that become accessible following phosphorylation at Ser172 can specifically detect the active form .

Advanced techniques like hydrogen-deuterium exchange mass spectrometry (HDX-MS) combined with epitope-specific antibodies can map conformational changes in TBK1 under different conditions. Proximity-based assays using conformation-sensitive antibodies can detect structural changes in living cells. For researchers developing conformation-specific antibodies, phage display libraries screened against specific conformational states of TBK1 can yield highly selective antibodies. When validating conformation-specific antibodies, it's essential to use both activating stimuli (like poly(I:C) treatment) and inhibitory conditions to confirm specificity. Structural data from techniques like cryo-EM or X-ray crystallography can further validate and explain the binding properties of conformation-specific antibodies. This approach requires rigorous validation but offers powerful insights into TBK1 activation dynamics that traditional antibody approaches cannot provide .

What strategies can address cross-reactivity issues when studying TBK1 among related kinases?

Addressing cross-reactivity issues when studying TBK1 among related kinases, particularly IKKε which shares significant homology, requires strategic approaches to ensure specificity. Epitope-targeted antibodies generated against unique regions of TBK1 that differ from related kinases can minimize cross-reactivity. These unique regions are typically found outside the highly conserved kinase domain. Simultaneous knockout validation using both TBK1 and related kinase (e.g., IKKε) knockout cell lines provides the most rigorous specificity testing. Researchers should test antibodies under both basal and stimulated conditions, as expression levels of kinases may change upon stimulation, potentially revealing cross-reactivity not apparent under basal conditions .

How do I design experiments to evaluate TBK1's role in disease models using antibody-based approaches?

Designing experiments to evaluate TBK1's role in disease models requires a multifaceted approach combining genetic, pharmacological, and antibody-based methods. Start by selecting appropriate disease models where TBK1 has been implicated, such as neurodegenerative disease models for ALS/FTD studies, or innate immunity models for infectious disease research. For neurodegenerative disease studies, consider both in vitro models (primary neurons, patient-derived iPSCs) and in vivo models (transgenic mice carrying TBK1 mutations). Establish proper controls including wild-type, heterozygous, and homozygous TBK1 knockout/mutant models to assess dose-dependent effects .

What are the best approaches for quantitative analysis of TBK1 expression and activation?

Quantitative analysis of TBK1 expression and activation requires precise methodologies to ensure accurate and reproducible results. For absolute quantification of TBK1 protein levels, quantitative Western blotting using recombinant TBK1 protein standards of known concentrations creates a calibration curve for sample measurement. Digital droplet PCR or qPCR with validated primers provides precise quantification of TBK1 mRNA expression. For activation assessment, the ratio of phosphorylated TBK1 (p-TBK1) to total TBK1 measured by Western blotting with phospho-specific and total antibodies serves as a key indicator of activation status .

ELISA-based approaches offer higher throughput quantification of both total and phosphorylated TBK1, while proximity ligation assays (PLA) enable quantitative in situ detection of TBK1 activation state in intact cells or tissues. Kinase activity assays using specific TBK1 substrates provide functional quantification of activation. When analyzing samples, researchers should normalize TBK1 expression to appropriate housekeeping proteins or total protein content (measured by methods like Ponceau S staining) to account for loading variations. For imaging-based quantification, automated image analysis platforms with machine learning algorithms can extract quantitative data on TBK1 expression and subcellular distribution from immunofluorescence images. Throughout these approaches, researchers should employ rigorous statistical analysis and report both biological and technical replicates to ensure robust quantification .

How can TBK1 antibodies be integrated with other research tools for comprehensive pathway analysis?

Integrating TBK1 antibodies with complementary research tools creates synergistic approaches for comprehensive pathway analysis. Combining immunoprecipitation using TBK1 antibodies with mass spectrometry (IP-MS) identifies TBK1 interactors and post-translational modifications under different conditions. ChIP-seq using TBK1 antibodies maps TBK1 associations with chromatin, revealing its role in transcriptional regulation. For dynamic studies, live-cell imaging with fluorescently tagged TBK1 antibody fragments (such as Fabs) can track endogenous TBK1 movement and localization in real time .

Multiplexed approaches are particularly powerful, such as combining TBK1 antibodies with antibodies against pathway components (IRF3, STING, etc.) in multi-color immunofluorescence or multiplex Western blotting to visualize pathway status. Integrating genetic tools (CRISPR-Cas9, RNAi) with antibody-based detection creates genetic-proteomic approaches to establish causality in pathway regulation. Pharmacological manipulations paired with antibody detection assess drug effects on TBK1 pathways. Computational biology tools can integrate antibody-based data with transcriptomics and proteomics datasets for systems-level pathway analysis. When using integrated approaches, researchers should validate that individual methods don't interfere with each other and carefully control for potential artifacts introduced by combining techniques. This integrated strategy provides a more complete understanding of TBK1's dynamic functions in complex cellular pathways than any single approach alone .

What considerations are important when selecting TBK1 antibodies for phosphorylation studies?

Phosphorylation studies of TBK1 require particularly careful antibody selection due to the critical role of phosphorylation in TBK1 activation and function. When selecting phospho-specific antibodies for TBK1, researchers should prioritize antibodies targeting Ser172 in the activation loop, which is the primary phosphorylation site indicating TBK1 activation. Monoclonal antibodies generally offer higher specificity for phospho-epitopes compared to polyclonal alternatives. Rigorous validation of phospho-specific antibodies should include demonstration of signal loss after phosphatase treatment and signal increase after pathway stimulation .

How to troubleshoot inconsistent results with TBK1 antibodies across different experimental conditions?

Inconsistent results with TBK1 antibodies can stem from multiple factors that require systematic troubleshooting. Begin by assessing antibody variability – lot-to-lot variations can significantly impact performance, so maintaining detailed records of antibody lots and standardizing to reference samples helps identify this issue. Cell type-specific differences in TBK1 expression levels, isoforms, or post-translational modifications can affect antibody recognition. Testing the antibody across multiple cell types with known TBK1 expression patterns can reveal such dependencies. Protocol inconsistencies, particularly in fixation methods for immunofluorescence or lysis conditions for Western blotting, can dramatically alter results. Standardizing protocols and documenting all steps in detail enhances reproducibility .

TBK1's phosphorylation state changes rapidly in response to cellular stimuli, so timing differences in sample collection can cause apparent inconsistencies. Standardizing the time between stimulation and sample collection is crucial. For challenging samples, epitope retrieval optimization (for tissue sections) or extraction method adjustments (for difficult-to-lyse samples) may be necessary. When inconsistencies persist, using multiple TBK1 antibodies targeting different epitopes can determine if the issue is epitope-specific. For comprehensive troubleshooting, consider orthogonal methods of detection not relying on antibodies, such as targeted mass spectrometry or genetic tagging approaches. Finally, consulting with both antibody manufacturers and other laboratories using the same antibody can provide valuable insights into known issues and solutions for specific TBK1 antibodies .

What are the best practices for developing or customizing TBK1 antibodies for specialized research applications?

Developing or customizing TBK1 antibodies for specialized applications requires strategic planning and rigorous validation. For epitope selection, analyze TBK1's sequence and structure to identify regions unique to TBK1 (avoiding conserved kinase domains shared with IKKε) and accessible in the protein's native conformation. Target selection should align with the intended application – surface-exposed epitopes for native applications versus internal segments for denatured applications. When immunizing animals, consider using multiple formats of the antigen (peptides, recombinant domains, full-length protein) to diversify the antibody response. For monoclonal antibody development, screening should include all intended applications (Western blot, IP, IF) using both recombinant TBK1 and endogenous protein from relevant cell types .

For specialized needs like conformation-specific antibodies, design screening strategies that distinguish between active and inactive TBK1 conformations. Modification-specific antibodies (targeting phosphorylation, ubiquitination, etc.) require careful negative control screening against unmodified protein. When developing application-specific variants, consider engineering modifications like fragment generation (Fab, scFv) for live-cell imaging or cross-linking antibodies to beads for improved immunoprecipitation. Throughout development, validation using TBK1 knockout cells or tissues is essential. For customized conjugations (fluorophores, enzymes), verify that the conjugation process doesn't impair epitope recognition. Custom antibody development typically requires 4-6 months and significant resources, so thoroughly evaluate whether existing antibodies can be optimized before initiating new development .

How do you account for the influence of TBK1 post-translational modifications on antibody binding efficacy?

Post-translational modifications (PTMs) of TBK1 can significantly affect antibody recognition, requiring careful consideration in experimental design and data interpretation. Phosphorylation, particularly at Ser172 in the activation loop, can alter protein conformation and either expose or mask antibody epitopes. Researchers should determine whether their antibodies recognize TBK1 regardless of phosphorylation status (phospho-independent) or are affected by phosphorylation. This can be tested by comparing antibody binding before and after phosphatase treatment. Ubiquitination of TBK1, important in its regulation, may interfere with antibody binding if the epitope includes or is adjacent to ubiquitination sites. Similarly, other modifications like SUMOylation or acetylation can affect antibody recognition .

To account for these influences, researchers should use multiple antibodies targeting different regions of TBK1 when possible. For critical experiments, pre-treating samples to remove specific modifications (using phosphatases, deubiquitinases, etc.) can help determine modification dependency. When analyzing TBK1 under conditions that alter its modification state (e.g., stimulation with pathogen-associated molecular patterns), consider how these changes might affect antibody binding. Mass spectrometry analysis of immunoprecipitated TBK1 can identify modifications present in your experimental system, helping interpret antibody binding variations. Additionally, researchers should clearly report the conditions under which antibodies were validated and acknowledge potential PTM-dependent recognition as a limitation in research publications. This comprehensive approach ensures accurate interpretation of TBK1 detection across different physiological and experimental conditions .

What are the considerations for using TBK1 antibodies in multiparameter and high-throughput screening approaches?

Using TBK1 antibodies in multiparameter and high-throughput screening approaches requires optimization beyond standard applications. For multiplexed immunofluorescence, select TBK1 antibodies with minimal spectral overlap with other fluorophores in your panel and verify that antibody performance isn't compromised in multiplexed settings through single-stain controls. When using TBK1 antibodies in flow cytometry, optimize fixation and permeabilization protocols specifically for intracellular kinase detection, as standard protocols may be insufficient. For automated imaging platforms, validate that TBK1 staining patterns are consistently detectable by your image analysis algorithms across various expression levels and treatment conditions .

In high-throughput ELISA or bead-based assays, determine the linear detection range of your TBK1 antibody to ensure quantitative accuracy and optimize antibody concentrations to maximize signal-to-noise ratio while minimizing consumption. For multiparameter analyses combining TBK1 with other biomarkers, verify that extraction or fixation conditions optimal for TBK1 detection don't compromise detection of other targets. In complex matrices like tissue lysates or biofluids, validate specificity using appropriate knockout controls in matrices matching your experimental samples. Implement robust quality control measures including positive and negative controls on each plate or batch. For large-scale screens, consider creating standardized positive control samples (e.g., lysates from stimulated cells with known TBK1 activation) that can be included across multiple batches to normalize inter-assay variation. Following these considerations ensures reliable TBK1 detection in complex and high-throughput experimental settings .

How are TBK1 antibodies being used in neurodegenerative disease research?

TBK1 antibodies have become increasingly important tools in neurodegenerative disease research due to the discovery of TBK1 mutations in amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). In clinical studies, TBK1 antibodies enable immunohistochemical analysis of patient tissues to examine TBK1 expression, localization, and aggregation patterns in affected brain regions. Phospho-specific TBK1 antibodies reveal activation status differences between normal and diseased states. In cellular models, TBK1 antibodies help investigate how disease-associated mutations affect TBK1 protein stability, kinase activity, and interaction with binding partners like OPTN (optineurin) and p62, which are critical for mitophagy and autophagy processes implicated in neurodegeneration .

Researchers are using TBK1 antibodies in combination with markers of autophagy, mitophagy, and inflammation to elucidate how TBK1 dysfunction contributes to disease pathogenesis. Co-immunoprecipitation with TBK1 antibodies followed by mass spectrometry identifies novel TBK1 interactors that may be disrupted in disease states. In animal models carrying TBK1 mutations, antibody-based approaches track changes in TBK1 distribution and function across disease progression. A particularly promising application involves using TBK1 antibodies to evaluate potential therapeutic interventions targeting the TBK1 pathway, including monitoring target engagement and pathway modulation in response to experimental drugs. As research progresses, multiparameter approaches combining TBK1 antibodies with other disease markers in single-cell analyses are providing unprecedented insights into cell type-specific roles of TBK1 in neurodegenerative processes .

What role do TBK1 antibodies play in studying innate immune responses and viral infections?

TBK1 antibodies are instrumental in studying innate immune responses and viral infections due to TBK1's critical role in type I interferon production. During viral infection studies, phospho-specific TBK1 antibodies track activation kinetics in response to various viral pathogens, including SARS-CoV-2, revealing how different viruses trigger or evade innate immune signaling. Immunofluorescence with TBK1 antibodies shows dynamic changes in subcellular localization during infection, particularly translocation to signaling complexes containing STING, MAVS, or other adaptor proteins. Co-immunoprecipitation studies using TBK1 antibodies identify virus-specific changes in TBK1 protein interactions, revealing how pathogens may subvert TBK1 signaling .

In broader innate immunity research, TBK1 antibodies help define the activation sequence in various pathogen recognition receptor pathways, including cGAS-STING, RIG-I-MAVS, and TLR signaling. Researchers combine TBK1 detection with measurements of downstream effectors like phosphorylated IRF3 and IFN-β production to map signaling cascades. Cell type-specific analyses using TBK1 antibodies reveal differential roles of TBK1 across immune cell populations such as macrophages, dendritic cells, and non-immune cells during infection. In vaccine research, particularly for COVID-19, TBK1 antibodies help characterize innate immune activation by vaccine components and breakthrough infections. These applications are contributing to our understanding of how TBK1 functions as a critical node in antiviral defense, with implications for developing immunomodulatory therapies for infectious and inflammatory diseases .

How can TBK1 antibodies contribute to cancer research and potential therapeutic development?

TBK1 antibodies serve as valuable tools in cancer research due to TBK1's emerging roles in oncogenic signaling, cell survival, and tumor microenvironment modulation. In tumor specimen analysis, TBK1 antibodies enable immunohistochemical profiling of TBK1 expression and activation across cancer types and stages, with some studies correlating high TBK1 expression or activation with poor prognosis. Phospho-specific TBK1 antibodies reveal hyperactivation of TBK1 in certain cancers, particularly those with KRAS mutations or STING pathway alterations. In mechanistic studies, co-immunoprecipitation with TBK1 antibodies identifies cancer-specific interaction partners that may drive oncogenic signaling or represent therapeutic vulnerabilities .

For studying tumor microenvironments, multiplexed immunofluorescence combining TBK1 antibodies with immune cell markers helps characterize TBK1's role in tumor-associated inflammation and anti-tumor immunity. In drug development contexts, TBK1 antibodies serve as essential tools for target validation, pharmacodynamic biomarker assessment, and mechanism of action studies for TBK1 inhibitors under investigation as cancer therapeutics. Researchers use TBK1 antibodies to evaluate combination therapies, particularly those combining TBK1 inhibition with immunotherapies or targeted agents. In patient-derived xenograft models, TBK1 antibodies help identify tumors likely to respond to TBK1-targeted therapies through analysis of baseline TBK1 activation. These diverse applications underscore how TBK1 antibodies contribute to understanding TBK1's complex roles in cancer biology and to developing novel therapeutic strategies targeting this kinase in appropriate cancer contexts .

How might advanced antibody engineering approaches improve TBK1 research tools?

Advanced antibody engineering holds significant promise for developing next-generation TBK1 research tools with enhanced capabilities. Single-domain antibodies (nanobodies) derived from camelid antibodies offer smaller size and superior tissue penetration compared to conventional antibodies, making them valuable for intracellular TBK1 targeting and super-resolution microscopy. Bispecific antibodies simultaneously targeting TBK1 and its binding partners (like STING or OPTN) could enable specific detection of functional TBK1 complexes rather than total TBK1 protein. Proximity-sensing antibody formats such as split nanoluciferase complementation systems can be engineered to produce signals only when TBK1 interacts with specific partners, enabling real-time monitoring of TBK1 interactions in living cells .

For monitoring TBK1 conformational changes, switchable antibody designs that produce detectable signals upon binding to activated TBK1 could revolutionize dynamic activity studies. Cell-permeable antibody formats would enable tracking of endogenous TBK1 in living cells without genetic modification. Using computational antibody design and directed evolution approaches could yield TBK1 antibodies with unprecedented specificity and affinity, particularly for challenging applications like distinguishing between closely related kinases. Additionally, site-specific conjugation technologies can produce homogeneous antibody-reporter conjugates with optimal orientation for detecting TBK1 in complex samples. These engineering approaches could transform TBK1 research by providing tools capable of answering questions about TBK1 dynamics, interactions, and functions that are currently difficult to address with conventional antibodies .

What are the prospects for integrating TBK1 antibodies with emerging single-cell and spatial analysis technologies?

The integration of TBK1 antibodies with emerging single-cell and spatial analysis technologies offers exciting prospects for understanding TBK1 biology with unprecedented resolution. In single-cell proteomics, TBK1 antibodies compatible with mass cytometry (CyTOF) or multiplexed ion beam imaging (MIBI) can reveal TBK1 expression and activation heterogeneity across thousands of individual cells within complex tissues. Spatial transcriptomics combined with TBK1 immunofluorescence creates multimodal datasets linking TBK1 protein expression with the broader transcriptional landscape at specific tissue locations. For in situ analysis of TBK1 interactions, proximity ligation assays with TBK1 antibodies can map protein-protein interactions within their native tissue context .

Advanced imaging technologies like multiplexed immunofluorescence using cyclic staining or DNA-barcoded antibodies can place TBK1 within the broader signaling architecture of cells in intact tissues. Imaging mass cytometry with metal-conjugated TBK1 antibodies enables simultaneous detection of dozens of proteins alongside TBK1 with subcellular resolution. For live tissues, intravital microscopy using fluorescently labeled TBK1 antibody fragments can track TBK1 dynamics in real time. These technologies require specifically optimized TBK1 antibodies and careful validation in the context of each platform, but they promise to transform our understanding of how TBK1 functions within the spatial organization of cells and tissues in both normal physiology and disease states. The integration of computational analysis, particularly machine learning approaches, with these antibody-based technologies will further enhance the extraction of biological insights from these complex, high-dimensional datasets .

How could TBK1 antibodies be utilized in developing clinical biomarkers for disease diagnosis or treatment response?

TBK1 antibodies hold significant potential for developing clinical biomarkers across several disease contexts. In neurodegenerative diseases like ALS and FTD, particularly for patients with TBK1 mutations, measuring TBK1 protein levels or phosphorylation status in cerebrospinal fluid or blood using highly sensitive immunoassays could provide diagnostic or prognostic information. Multiplex immunoassays incorporating TBK1 antibodies alongside other disease markers might improve diagnostic accuracy or patient stratification. For cancer applications, TBK1 activation status assessed by phospho-specific antibodies in tumor biopsies could potentially predict response to therapies targeting STING, KRAS, or other pathways intersecting with TBK1 signaling .

In the realm of inflammatory and autoimmune diseases, where TBK1 plays important roles in innate immune signaling, monitoring TBK1 activation in peripheral blood immune cells using phospho-flow cytometry with TBK1 antibodies might track disease activity or treatment response. For developing clinical applications, technological platforms like automated immunohistochemistry systems or bead-based multiplex assays incorporating validated TBK1 antibodies would enable standardized testing in clinical laboratories. The key challenges for clinical translation include developing antibody-based assays with sufficient sensitivity, specificity, and reproducibility for detecting TBK1 in clinical specimens, as well as establishing clear associations between TBK1 measurements and clinically meaningful outcomes through well-designed clinical studies. As TBK1's roles in various diseases become better defined, antibody-based biomarker approaches targeting this kinase may contribute to precision medicine approaches for patient selection and treatment monitoring .

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.