TTC14 Antibody, Biotin conjugated

Shipped with Ice Packs
In Stock

Description

Target Protein: TTC14

TTC14 (Tetratricopeptide repeat protein 14) is a 770-amino-acid protein containing tetratricopeptide (TPR) repeats, which mediate protein-protein interactions. It is implicated in cellular processes such as transcriptional regulation and mitochondrial function .

Key Features of TTC14:

  • Gene ID: 151613

  • Molecular Weight: ~88 kDa (predicted)

  • Expression: Ubiquitous, with roles in testis, skin, and intestinal epithelium .

Biotin Conjugation Mechanism

Biotinylation enables high-affinity binding to streptavidin (K<sub>d</sub> ~10<sup>−14</sup> M), facilitating signal amplification in assays . This antibody uses covalent linkage of biotin to lysine residues or Fc regions, preserving antigen-binding specificity .

Advantages of Biotinylation:

  • Enhanced detection sensitivity in multiplex assays.

  • Compatibility with streptavidin-HRP or fluorophore conjugates.

  • Reduced nonspecific binding compared to traditional labeling methods .

4.1. Immunoassays

  • ELISA: Used for quantitative detection of TTC14 in serum or cell lysates .

  • Western Blot: Validated in Jurkat, Daudi, and Ramos cell lines with a predicted 88 kDa band .

  • Flow Cytometry: Detects intracellular TTC14 in permeabilized cells .

4.2. Advanced Techniques

  • Proximity Labeling: Biotinylated antibodies enable spatial proteomics via APEX peroxidase systems .

  • Antibody-Drug Conjugate (ADC) Screening: Streptavidin-biotin platforms allow rapid evaluation of toxin-antibody pairs for cancer therapy .

Comparative Research Findings

  • Sensitivity: Anti-biotin antibodies achieve 30-fold higher biotinylated peptide enrichment than streptavidin-based methods .

  • Cross-Reactivity: Polyclonal TTC14 antibodies show high specificity for human samples, with no observed cross-reactivity in non-human models .

  • Therapeutic Potential: Biotinylated antibodies like Trastuzumab-SB-DM1 demonstrate efficacy comparable to FDA-approved ADCs in preclinical models .

Limitations

  • Batch Variability: Polyclonal antibodies may exhibit lot-to-lot variability .

  • Application Restrictions: Not all conjugates are validated for in vivo use due to ProClin toxicity .

Future Directions

Ongoing research focuses on optimizing biotinylation sites to minimize interference with antigen-binding domains and expanding applications in spatial transcriptomics .

Product Specs

Buffer
**Preservative:** 0.03% Proclin 300
**Constituents:** 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship products within 1-3 business days of receiving your order. Delivery times may vary based on the chosen shipping method and location. Please contact your local distributor for specific delivery timeframes.
Synonyms
Tetratricopeptide repeat protein 14 (TPR repeat protein 14), TTC14, KIAA1980
Target Names
TTC14
Uniprot No.

Q&A

What is TTC14 protein and what is its biological significance?

Tetratricopeptide repeat protein 14 (TTC14) is a human protein characterized by the presence of tetratricopeptide repeat domains, which are structural motifs consisting of 34 amino acid repeats that mediate protein-protein interactions. TTC14 is encoded by the TTC14 gene (also known as KIAA1980 or UNQ5813/PRO19630) and is identified in the UniProt database with the primary accession number Q96N46 . The protein has a predicted molecular weight of approximately 88 kDa and is expressed in various human cell lines including Daudi, Ramos, and Jurkat cells, suggesting potential roles in immune cell function . While the complete functional characterization of TTC14 remains an active area of research, its structural motifs suggest involvement in protein complex assembly, protein transport, and possibly cell cycle regulation, making it a valuable target for investigation in fundamental cellular biology.

What is the significance of biotin conjugation in TTC14 antibodies for research applications?

Biotin conjugation of TTC14 antibodies represents a strategic modification that significantly enhances detection versatility through the exploitation of the exceptionally high-affinity interaction between biotin and streptavidin (Kd ≈ 10^-15 M). This conjugation enables several methodological advantages in experimental systems. The biotin-conjugated TTC14 antibody can be used with various streptavidin-conjugated detection systems (HRP, fluorophores, gold particles), allowing for signal amplification and increased detection sensitivity in ELISA and other immunoassays . Additionally, the small size of biotin (244 Da) minimizes steric hindrance that might otherwise interfere with antibody-antigen binding, preserving the recognition specificity for TTC14 protein epitopes. The commercially available biotin-conjugated TTC14 antibody is derived from rabbit hosts and demonstrates reactivity with human TTC14, particularly with epitope regions between amino acids 569-770 .

How do polyclonal and monoclonal TTC14 antibodies differ in research applications?

Both polyclonal and monoclonal TTC14 antibodies are available for research, each offering distinct advantages depending on experimental requirements:

FeaturePolyclonal TTC14 AntibodiesMonoclonal TTC14 Antibodies
SourceGenerated in rabbits against recombinant TTC14 protein fragmentsProduced from single B-cell clones (e.g., EPR11143 clone)
Epitope recognitionRecognize multiple epitopes on TTC14 (e.g., regions 569-770 AA) Target single, specific epitope on TTC14
Signal strengthGenerally stronger signal due to multiple epitope bindingMay have lower sensitivity but higher specificity
Lot-to-lot variabilityHigher variability between production batchesConsistent performance across batches
Best applicationsELISA, immunofluorescence, initial protein characterizationWestern blotting, flow cytometry, applications requiring reproducible results
Cross-reactivityPotentially higher cross-reactivity with related proteinsTypically lower cross-reactivity

When selecting between these antibody types, researchers should consider whether signal strength (favoring polyclonal) or specificity and reproducibility (favoring monoclonal) is the higher priority for their experimental design .

What are the optimal validated applications for biotin-conjugated TTC14 antibodies?

Based on empirical validation, biotin-conjugated TTC14 antibodies have been specifically tested and optimized for enzyme-linked immunosorbent assay (ELISA) applications . The biotin conjugation is particularly advantageous in ELISA formats as it enables signal amplification through streptavidin-coupled detection systems. While ELISA represents the primary validated application, the versatility of biotin-conjugated antibodies suggests potential utility in other techniques where a streptavidin-based detection system can be employed. These might include immunohistochemistry, immunocytochemistry, and potentially protein microarray applications, though researchers should conduct appropriate validation for these secondary applications. When designing experiments with biotin-conjugated TTC14 antibodies, it is recommended to begin with ELISA protocols where established performance parameters are available, before extending to alternative methodologies .

What are the optimal storage and handling conditions for maintaining biotin-conjugated TTC14 antibody activity?

To preserve the functional integrity of biotin-conjugated TTC14 antibodies, strict adherence to proper storage and handling protocols is essential:

  • Storage temperature: Maintain at -20°C for long-term storage .

  • Aliquoting: Upon receipt, divide the antibody into small, single-use aliquots to minimize freeze-thaw cycles .

  • Freeze-thaw cycles: Strictly limit repeated freezing and thawing as this can lead to denaturation and loss of binding activity .

  • Storage buffer: The antibody is typically supplied in 0.01 M PBS, pH 7.4, with 0.03% Proclin-300 and 50% glycerol, which helps stabilize the protein during freeze-thaw transitions .

  • Working dilutions: Prepare immediately before use and do not store diluted antibody for extended periods.

  • Light exposure: Minimize exposure to light, particularly important for biotin conjugates to prevent photobleaching of the biotin moiety.

Researchers should monitor antibody performance regularly when using stored antibodies, as degradation can occur even under optimal storage conditions, potentially leading to reduced signal intensity or increased background .

How should researchers determine the optimal working dilution for biotin-conjugated TTC14 antibodies?

Determining the optimal working dilution for biotin-conjugated TTC14 antibodies requires a systematic titration approach to balance specific signal detection with minimal background. While manufacturer guidelines suggest that "optimal dilutions/concentrations should be determined by the end user" , a methodical approach should follow these steps:

  • Initial range finding: Begin with a broad dilution series (e.g., 1:100, 1:500, 1:1000, 1:5000) using positive controls (samples known to express TTC14) and negative controls.

  • Signal-to-noise optimization: Calculate the signal-to-noise ratio for each dilution by dividing the positive control signal by the negative control signal.

  • Fine titration: Narrow the dilution range around the concentration showing the highest signal-to-noise ratio and repeat with smaller increments.

  • Cross-validation: Verify the selected dilution across multiple sample types relevant to your research.

  • Lot-to-lot validation: When receiving a new antibody lot, perform abbreviated titration to confirm optimal dilution.

This empirical approach is essential because optimal concentrations vary depending on the specific application, sample type, detection system, and individual laboratory conditions. Documentation of these optimization steps strengthens experimental reproducibility and is recommended for inclusion in materials and methods sections of publications .

What controls are essential when using biotin-conjugated TTC14 antibodies in experimental systems?

Implementing a comprehensive set of controls is crucial for ensuring experimental validity and interpretability when working with biotin-conjugated TTC14 antibodies:

Control TypePurposeImplementation
Positive ControlConfirms antibody functionalityUse cell lines with confirmed TTC14 expression (e.g., Daudi, Ramos, or Jurkat cells)
Negative ControlEstablishes background signal levelsUse cell lines lacking TTC14 expression or TTC14 knockout samples
Isotype ControlDistinguishes non-specific bindingInclude rabbit IgG (matching isotype) at equivalent concentration
Secondary-only ControlIdentifies background from detection systemOmit primary antibody but include streptavidin detection reagent
Biotin Blocking ControlAssesses endogenous biotin interferencePre-block samples with streptavidin before adding biotin-conjugated antibody
Peptide CompetitionConfirms epitope specificityPre-incubate antibody with recombinant TTC14 protein (569-770 AA region)
Antibody TitrationOptimizes signal-to-noise ratioTest serial dilutions to identify optimal concentration

These controls should be systematically incorporated into experimental design and collectively provide a framework for distinguishing genuine TTC14 detection from technical artifacts or non-specific signals .

How does biotin supplementation potentially interfere with assays using biotin-conjugated TTC14 antibodies?

Biotin supplementation presents a significant confounding factor in assays utilizing biotin-conjugated antibodies through competitive binding mechanisms. When biological samples contain elevated biotin levels—common with subjects taking biotin supplements—this free biotin competes with biotin-conjugated TTC14 antibodies for binding to streptavidin in detection systems. This competition can manifest in several ways:

  • Signal suppression: High concentrations of free biotin can displace biotin-conjugated antibodies from streptavidin detection reagents, resulting in falsely decreased signal intensity.

  • Variable interference: The effect varies based on biotin concentration in the sample. Standard multivitamin preparations (~30 μg biotin) typically cause minimal interference, but specialized supplements can contain up to 650 times the recommended daily intake .

  • Elimination kinetics: Biotin has a dose-dependent elimination half-life—higher doses (>30 mg/day) may require several days to clear sufficiently from samples .

To mitigate biotin interference when working with clinical samples:

  • Document any biotin supplementation in study subjects

  • Consider a biotin-free washout period before sample collection (8-72 hours depending on biotin dose)

  • Implement streptavidin pre-blocking steps in sample preparation

  • Include biotin-spiked controls to quantify potential interference levels

These considerations are particularly important in translational research contexts where samples might come from subjects taking biotin supplements .

What are common troubleshooting approaches for poor signal detection with biotin-conjugated TTC14 antibodies?

When encountering suboptimal signal detection with biotin-conjugated TTC14 antibodies, systematic troubleshooting should address multiple potential causes:

  • Antibody functionality issues:

    • Verify antibody hasn't undergone excessive freeze-thaw cycles

    • Check storage conditions (maintain at -20°C, avoid prolonged room temperature exposure)

    • Confirm antibody hasn't exceeded recommended shelf life

    • Test a fresh aliquot from a different lot if available

  • Sample preparation factors:

    • Ensure appropriate protein extraction methods preserving TTC14 epitopes

    • Verify protein concentration is sufficient for detection

    • Consider epitope masking due to protein folding or post-translational modifications

    • Test different sample preparation buffers if native conformation is required

  • Detection system optimization:

    • Increase streptavidin-conjugate concentration

    • Extend incubation times for both primary antibody and detection reagent

    • Verify streptavidin reagent functionality with a biotin standard

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

  • Endogenous biotin interference:

    • Implement avidin/streptavidin blocking steps before antibody application

    • Use streptavidin variants with reduced sensitivity to free biotin

    • Consider switching to non-biotin detection systems if interference persists

  • Application-specific adjustments:

    • For ELISA: Optimize coating buffer, blocking reagents, and washing stringency

    • For immunofluorescence: Test different fixation methods (paraformaldehyde vs. methanol)

    • For Western blotting: Verify transfer efficiency and membrane blocking protocol

Systematic evaluation of these factors, changing one variable at a time while maintaining appropriate controls, will help identify and resolve the source of poor signal detection .

How can researchers effectively validate the specificity of TTC14 antibody binding in their experimental systems?

Comprehensive validation of TTC14 antibody specificity is essential for ensuring experimental rigor and reproducibility. A multi-faceted validation approach should incorporate the following methodologies:

  • Genetic validation:

    • Use TTC14 knockout or knockdown models (CRISPR-Cas9, RNAi) to confirm signal disappearance

    • Employ TTC14 overexpression systems to demonstrate signal enhancement

    • Compare signal across genetically diverse cell lines with known TTC14 expression profiles

  • Molecular validation:

    • Perform peptide competition assays using the immunizing peptide (569-770 AA region for many commercial antibodies)

    • Compare multiple antibodies targeting different TTC14 epitopes

    • Confirm molecular weight concordance (expected 88 kDa) in Western blot applications

  • Orthogonal method confirmation:

    • Correlate antibody-based detection with mRNA expression (qPCR or RNA-seq)

    • Use mass spectrometry to confirm protein identity in immunoprecipitated samples

    • Employ proximity ligation assays to verify interaction with known binding partners

  • Cross-reactivity assessment:

    • Test antibody against related tetratricopeptide repeat proteins

    • Evaluate species cross-reactivity if working with non-human models

    • Address potential cross-reactivity with proteins containing similar structural motifs

  • Technical validation:

    • Compare signal patterns across multiple applications (ELISA, Western blot, immunofluorescence)

    • Document reproducibility across different lots of the same antibody

    • Verify signal linearity across a range of protein concentrations

This comprehensive validation framework significantly strengthens confidence in experimental findings and should be documented in publications to enhance research reproducibility .

What are the current advanced applications of TTC14 antibodies in multiplex immunoassay systems?

Multiplex immunoassay platforms represent an evolving frontier for TTC14 antibody applications, offering simultaneous detection of multiple analytes to provide contextual understanding of TTC14 in complex biological systems. Integration of biotin-conjugated TTC14 antibodies into these platforms requires specific methodological considerations:

  • Bead-based multiplex systems:

    • TTC14 antibodies can be conjugated to spectrally distinct beads alongside antibodies for interacting partners

    • Differential streptavidin-fluorophore conjugates enable simultaneous detection of multiple biotin-labeled antibodies

    • Critical consideration: careful titration to prevent cross-platform interference

  • Protein microarray integration:

    • TTC14 antibodies can be spotted alongside antibodies against related tetratricopeptide repeat proteins

    • Enables comparative binding analysis across protein families

    • Implementation challenge: maintaining consistent binding conditions across diverse antibodies

  • Sequential multiplex immunohistochemistry:

    • Tyramide signal amplification with spectral unmixing allows detection of TTC14 alongside multiple markers

    • Particularly valuable for spatial context analysis in tissue samples

    • Technical requirement: optimization of antibody stripping protocols between rounds

  • Single-cell proteomics platforms:

    • Integration with mass cytometry (CyTOF) using metal-tagged streptavidin

    • Enables correlation of TTC14 expression with dozens of cellular markers

    • Methodological consideration: careful panel design to avoid signal spillover

  • Proximity-based detection systems:

    • Combination with proximity ligation or proximity extension assays

    • Allows detection of TTC14 interactions with specific partner proteins

    • Advanced application: verification of protein complex formation in native conditions

These multiplex approaches facilitate systems-level analysis of TTC14 biology, though each requires specific optimization beyond standard single-plex applications .

How should researchers integrate TTC14 antibody data with other -omics approaches for comprehensive biological insights?

Integration of TTC14 antibody-generated data with complementary -omics methodologies creates opportunities for multi-dimensional biological insights through the following structured approaches:

  • Correlation with transcriptomics:

    • Compare TTC14 protein levels (antibody-detected) with TTC14 mRNA expression

    • Investigate potential post-transcriptional regulation when protein-mRNA correlations diverge

    • Methodological approach: Develop normalization strategies that account for different dynamic ranges

    • Case application: Identify cellular contexts where TTC14 protein stability may be regulated independently of transcription

  • Integration with interactomics:

    • Use TTC14 antibodies for immunoprecipitation followed by mass spectrometry

    • Map TTC14 protein-protein interaction networks under different cellular conditions

    • Technical consideration: Validate that antibody binding doesn't disrupt native protein interactions

    • Research opportunity: Characterize the complete interactome of TTC14 to elucidate its functional roles

  • Correlation with phosphoproteomics:

    • Combine TTC14 detection with phospho-specific antibodies

    • Investigate how post-translational modifications affect TTC14 function

    • Implementation strategy: Develop sequential immunoprecipitation protocols using TTC14 antibodies followed by phospho-enrichment

    • Analytical approach: Time-course studies to map signaling dynamics

  • Integration with spatial -omics:

    • Use TTC14 antibodies in spatial transcriptomics or proteomics platforms

    • Map subcellular localization patterns in relation to other biomolecules

    • Technology application: Combine with emerging spatial multiomics platforms (e.g., 10x Visium with immunofluorescence)

    • Analysis focus: Identify microenvironmental factors influencing TTC14 localization

  • Computational integration frameworks:

    • Develop computational pipelines for meaningful integration of antibody-based quantification with other data types

    • Apply machine learning approaches to identify patterns across multi-omics datasets including TTC14

    • Methodological requirement: Standardized data normalization across different measurement platforms

    • Validation approach: Experimental testing of computationally predicted TTC14 functional relationships

This multi-omics integration strategy ultimately positions TTC14 antibody data within broader biological contexts, potentially revealing functional roles and regulatory mechanisms that would remain obscured through single-platform analysis .

What are the current limitations of available TTC14 antibodies and how might these be addressed in future research?

Current TTC14 antibodies present several limitations that constrain research applications, along with emerging strategies to address these challenges:

  • Epitope coverage limitations:

    • Current issue: Most commercial antibodies target specific regions (e.g., 569-770 AA or 470-664 AA) , potentially missing conformational epitopes

    • Future direction: Development of antibodies targeting diverse epitopes across the full TTC14 protein to enable comprehensive structural and functional analysis

    • Methodological advancement: Phage display technology to generate antibodies against challenging epitopes

  • Cross-reactivity concerns:

    • Current limitation: Potential cross-reactivity with other tetratricopeptide repeat proteins due to structural similarities

    • Emerging solution: Enhanced validation using CRISPR knockout cell lines to definitively establish specificity

    • Technical need: Systematic cross-reactivity testing against related protein family members

  • Species reactivity restrictions:

    • Present constraint: Primary reactivity limited to human TTC14 , limiting comparative studies

    • Future development: Generation of antibodies with validated cross-species reactivity to facilitate evolutionary and animal model research

    • Approach: Strategic immunogen design targeting conserved regions across species

  • Application versatility:

    • Current limitation: Primarily validated for ELISA and Western blot applications

    • Future direction: Comprehensive validation across broader technique spectrum including ChIP, super-resolution microscopy, and live-cell imaging

    • Technical advance: Site-specific conjugation methods to preserve antibody functionality

  • Quantitative standardization:

    • Present challenge: Lack of standardized quantification methods across laboratories

    • Emerging solution: Development of recombinant TTC14 reference standards for absolute quantification

    • Methodological improvement: Digital immunoassay platforms for higher sensitivity and dynamic range

These limitations represent opportunities for antibody engineering and validation advances that will ultimately expand the research utility of TTC14 antibodies .

How can researchers effectively design experiments to investigate TTC14 protein interactions using biotin-conjugated antibodies?

Designing robust experiments to elucidate TTC14 protein interactions requires strategic utilization of biotin-conjugated antibodies within carefully structured experimental frameworks:

  • Co-immunoprecipitation strategies:

    • Leverage biotin-conjugated TTC14 antibodies with streptavidin-coated magnetic beads for efficient pull-down

    • Implementation approach: Reverse co-IP validation where putative interaction partners are immunoprecipitated and probed for TTC14

    • Critical control: Include isotype-matched biotin-conjugated antibodies to identify non-specific binding

    • Technical consideration: Optimize lysis conditions to preserve native protein complexes

  • Proximity-based interaction detection:

    • Combine biotin-conjugated TTC14 antibodies with proximity ligation assay technology

    • Experimental design: Dual recognition approach requiring second antibody against candidate interaction partner

    • Analytical advantage: Provides spatial context of protein interactions within cellular compartments

    • Validation approach: Confirm interactions using genetic manipulation of putative partners

  • Competitive binding analysis:

    • Use biotin-conjugated TTC14 antibodies to investigate competition between different binding partners

    • Methodological approach: Pre-incubation with unlabeled potential competitors followed by TTC14 immunoprecipitation

    • Quantification strategy: Develop dose-response curves for competitive displacement

    • Control design: Include structurally related non-competitor proteins

  • Affinity measurement platforms:

    • Employ surface plasmon resonance or biolayer interferometry with immobilized biotin-TTC14 antibody complexes

    • Experimental setup: Capture TTC14 protein and measure binding kinetics with putative partners

    • Data analysis: Determine association/dissociation rate constants and equilibrium binding constants

    • Technical requirement: Careful surface regeneration between measurement cycles

  • Dynamic interaction analysis:

    • Utilize biotin-conjugated TTC14 antibodies in live-cell imaging applications with streptavidin-fluorophore labeling

    • Implementation approach: Microinjection of minimally disruptive antibody fragments

    • Technical innovation: Combine with optogenetic perturbation to trigger interaction events

    • Analysis framework: Quantitative tracking of molecular dynamics following stimulation

These experimental approaches, when implemented with appropriate controls and quantitative analysis, provide powerful frameworks for systematically characterizing the TTC14 interactome .

What are the potential research applications of TTC14 antibodies in understanding disease mechanisms?

While TTC14 research remains primarily in foundational investigative stages, biotin-conjugated TTC14 antibodies offer promising applications for elucidating potential disease associations through several methodological approaches:

  • Biomarker investigation in pathological samples:

    • Apply TTC14 antibodies in tissue microarray analysis spanning diverse pathologies

    • Methodological approach: Multiplex immunohistochemistry to correlate TTC14 expression with established disease markers

    • Analytical strategy: Quantitative image analysis to detect subtle expression pattern changes

    • Research opportunity: Correlation of TTC14 levels or localization with disease progression or therapeutic response

  • Functional analysis in disease models:

    • Deploy TTC14 antibodies to track protein dynamics in cellular disease models

    • Experimental design: Compare TTC14 interaction networks between normal and disease-state cells

    • Technical implementation: Combine with CRISPR-mediated TTC14 modification to assess causality

    • Validation approach: Rescue experiments to confirm specificity of observed phenotypes

  • Post-translational modification analysis in pathological conditions:

    • Combine TTC14 antibodies with modification-specific antibodies (phospho, ubiquitin, etc.)

    • Methodological framework: Sequential immunoprecipitation to enrich for modified TTC14 fractions

    • Analytical opportunity: Mass spectrometry characterization of disease-specific modifications

    • Research direction: Identification of modification-dependent interaction partners in disease contexts

  • Protein mislocalization investigation:

    • Utilize TTC14 antibodies for subcellular fractionation and imaging studies

    • Experimental approach: Compare TTC14 localization patterns between normal and pathological samples

    • Technical consideration: Super-resolution microscopy to detect subtle localization changes

    • Analytical framework: Correlation of mislocalization with functional consequences

  • Therapeutic target validation:

    • Apply TTC14 antibodies to validate target engagement in drug development pipelines

    • Methodological implementation: Competitive binding assays with candidate therapeutic compounds

    • Research application: Monitor TTC14 complex formation changes in response to experimental therapeutics

    • Translational opportunity: Development of proximity-based assays for high-throughput screening platforms

These research applications provide frameworks for investigating TTC14's potential roles in disease mechanisms, though comprehensive characterization would require integration with broader experimental approaches including genetic models and clinical correlation studies .

What are the current best practices for reporting TTC14 antibody-based research in scientific publications?

To ensure reproducibility and rigor in TTC14 antibody-based research, publications should adhere to comprehensive reporting standards that address multiple dimensions of antibody methodology:

  • Antibody identification and sourcing:

    • Report complete antibody identifiers including catalog number, clone ID if monoclonal, and lot number

    • Specify host species, clonality (polyclonal vs. monoclonal), and any conjugations (e.g., biotin)

    • Disclose commercial source with full company name and location

    • For custom antibodies, provide detailed immunogen information including the specific TTC14 region used (e.g., 569-770 AA)

  • Validation documentation:

    • Describe all validation steps performed (Western blot, peptide competition, knockout controls)

    • Include validation data in supplementary materials if not previously published

    • Reference prior publications establishing antibody specificity, if applicable

    • Disclose any known cross-reactivity or limitations

  • Methodology transparency:

    • Detail precise protocols including antibody dilutions, incubation times, temperatures, and buffers

    • Specify detection systems used with biotin-conjugated antibodies (e.g., streptavidin-HRP dilution)

    • Document any modifications to manufacturer's recommended protocols

    • Report replicate structure and statistical approaches for quantitative analyses

  • Control implementation:

    • Describe all controls employed (negative, positive, isotype, blocking)

    • Include representative images or data from controls in figures or supplements

    • Explain how controls informed data interpretation and troubleshooting

    • Disclose any control results that indicated potential limitations

  • Data availability:

    • Provide access to full, unprocessed immunoblot or microscopy images through data repositories

    • Deposit detailed protocols in repositories such as protocols.io

    • Share analysis code used for quantification of antibody-generated data

    • Enable reagent sharing through appropriate material transfer agreements

Adherence to these reporting standards enhances experimental reproducibility and accelerates collective progress in TTC14 research by enabling effective knowledge transfer between laboratories .

How can researchers integrate computational approaches with TTC14 antibody research for enhanced insights?

Computational approaches offer powerful complementary strategies to extend the insights gained from TTC14 antibody-based research through several methodological frameworks:

  • Structural prediction integration:

    • Combine epitope mapping data from TTC14 antibodies with protein structure prediction algorithms

    • Implementation approach: Use antibody accessibility information to refine computational models

    • Analytical workflow: Map antibody binding sites onto predicted 3D structures to infer functional domains

    • Research application: Guide rational design of functional experiments targeting specific structural features

  • Network biology approaches:

    • Integrate TTC14 interaction data from antibody-based experiments with protein-protein interaction databases

    • Computational strategy: Apply network analysis algorithms to identify potential functional modules

    • Technical implementation: Bayesian integration of antibody-derived interaction data with public databases

    • Research opportunity: Identification of previously unrecognized functional associations for experimental validation

  • Machine learning for image analysis:

    • Apply deep learning approaches to TTC14 immunofluorescence or immunohistochemistry images

    • Methodological framework: Train neural networks to recognize subtle pattern differences in TTC14 localization

    • Technical advantage: Detection of patterns not apparent through conventional visual inspection

    • Implementation strategy: Use transfer learning with pre-trained networks adapted to TTC14-specific features

  • Multi-omics data integration:

    • Develop computational pipelines to correlate TTC14 antibody-generated data with transcriptomic and proteomic datasets

    • Analytical approach: Apply dimension reduction techniques to identify coordinated changes across data types

    • Technical consideration: Implement robust normalization strategies across heterogeneous data platforms

    • Research application: Generate testable hypotheses about TTC14 regulation and function

  • Virtual screening approaches:

    • Use antibody-derived binding site information to inform computational screening for TTC14-targeting compounds

    • Implementation strategy: Develop docking models based on antibody epitope competition data

    • Technical opportunity: Leverage antibody competition assays to validate in silico predictions

    • Research direction: Structure-based design of tools to modulate TTC14 interactions

These computational approaches extend beyond what antibodies alone can achieve, creating synergistic frameworks for deeper understanding of TTC14 biology when integrated with experimental validation .

What standardized protocols are recommended for using biotin-conjugated TTC14 antibodies in ELISA applications?

The following standardized ELISA protocol is optimized for biotin-conjugated TTC14 antibodies, incorporating critical quality control steps and technical considerations:

Standard ELISA Protocol for Biotin-Conjugated TTC14 Antibody:

  • Plate preparation:

    • Coat high-binding 96-well plates with capture antibody (anti-TTC14) at 1-2 μg/mL in carbonate buffer (pH 9.6)

    • Incubate overnight at 4°C

    • Wash 3× with PBS containing 0.05% Tween-20 (PBST)

  • Blocking:

    • Block non-specific binding with 1-5% BSA in PBS for 1-2 hours at room temperature

    • Critical consideration: Use biotin-free BSA to prevent interference with detection system

    • Wash 3× with PBST

  • Sample application:

    • Apply cell lysates or purified protein samples diluted in blocking buffer

    • Include a standard curve using recombinant TTC14 protein (569-770 AA region)

    • Incubate 1-2 hours at room temperature or overnight at 4°C

    • Wash 4× with PBST

  • Primary antibody incubation:

    • Apply biotin-conjugated TTC14 antibody

    • Starting dilution recommendation: 1:1000 in blocking buffer (optimize through titration)

    • Incubate 1-2 hours at room temperature

    • Wash 5× with PBST

  • Detection:

    • Apply streptavidin-HRP at 1:5000 to 1:20000 dilution in blocking buffer

    • Incubate 30-60 minutes at room temperature

    • Wash 5× with PBST

  • Signal development:

    • Add TMB substrate and monitor color development

    • Stop reaction with 2N H₂SO₄ when appropriate signal-to-noise ratio is achieved

    • Read absorbance at 450 nm with 570 nm reference wavelength

  • Critical controls:

    • Include wells without sample (blank)

    • Include isotype control antibody (rabbit IgG-biotin)

    • Include free biotin competition control to assess specificity

    • Process all controls identically to experimental samples

  • Data analysis:

    • Subtract background (blank) values from all readings

    • Generate standard curve using four-parameter logistic regression

    • Determine sample concentrations from standard curve

    • Document and report all validation metrics including assay dynamic range, sensitivity, and variability

This standardized protocol provides a foundation for reliable TTC14 quantification while incorporating specific considerations for biotin-conjugated antibody applications .

What are the key technical differences when working with biotin-conjugated versus unconjugated TTC14 antibodies?

Working with biotin-conjugated TTC14 antibodies introduces several important technical distinctions compared to unconjugated formats, requiring specific methodological adaptations:

Technical AspectBiotin-Conjugated TTC14 AntibodiesUnconjugated TTC14 Antibodies
Detection systemRequires streptavidin-coupled reporter (HRP, fluorophore, etc.) Requires species-specific secondary antibody
Signal amplificationHigher sensitivity through avidin-biotin complex formationLimited to single secondary antibody binding per primary
Multiplexing capabilityCan be combined with antibodies of same species without cross-reactivitySpecies limitations in multiplexing applications
Endogenous biotin interferenceSusceptible to competition from endogenous biotin in samples No direct interference from endogenous biotin
Storage considerationsMore sensitive to freeze-thaw degradation of biotin moiety Generally more stable through freeze-thaw cycles
Working dilution rangeTypically used at higher dilutions due to signal amplificationOften requires lower dilutions for equivalent signal
Background issuesPotential background from endogenous biotin-binding proteinsBackground primarily from non-specific antibody binding
Blocking requirementsRequires biotin-free blocking reagents Standard blocking protocols apply
Fixation compatibilityMay have reduced performance with certain fixatives affecting biotinGenerally more compatible with diverse fixation methods
Application versatilityOptimal for ELISA, may have limitations in certain applications Broader application range including IP, ChIP, IHC

These technical differences necessitate specific protocol adaptations when transitioning between biotin-conjugated and unconjugated TTC14 antibodies, even when the base antibody is derived from the same clone or polyclonal source .

What quantitative approaches are recommended for analyzing TTC14 expression data generated with antibody-based methods?

Rigorous quantitative analysis of TTC14 antibody-generated data requires application of appropriate statistical and computational approaches tailored to specific experimental platforms:

  • Western blot densitometry:

    • Methodology: Normalize TTC14 band intensity to loading controls (β-actin, GAPDH)

    • Statistical approach: Apply log-transformation before parametric testing due to non-normal distribution

    • Quality control: Verify signal linearity across protein loading range (5-50 μg)

    • Validation requirement: Confirm quantification with multiple exposure times to avoid saturation

  • ELISA quantification:

    • Standard curve modeling: Use four-parameter logistic regression rather than linear interpolation

    • Technical validation: Calculate intra-assay and inter-assay coefficients of variation (<15% acceptable)

    • Statistical consideration: Account for plate effects through appropriate experimental design

    • Data reporting: Include lower limit of detection and quantification in methods

  • Immunofluorescence image analysis:

    • Quantification approach: Apply automated segmentation to define regions of interest

    • Parameter selection: Measure integrated intensity rather than maximum intensity

    • Reference normalization: Normalize to nuclear counterstain or cell surface area

    • Statistical requirement: Analyze sufficient cell numbers (>100 per condition) to account for heterogeneity

  • Flow cytometry data:

    • Gating strategy: Document consistent gating approach for TTC14-positive populations

    • Expression metrics: Report median fluorescence intensity rather than mean

    • Statistical analysis: Apply non-parametric methods for comparing distributions

    • Technical consideration: Include fluorescence-minus-one controls for accurate gate setting

  • Multiplex assay analysis:

    • Cross-assay normalization: Include reference standards across all assay runs

    • Data integration: Apply batch correction algorithms when combining datasets

    • Statistical approach: Use multivariate methods to identify correlated expression patterns

    • Validation strategy: Confirm key findings with orthogonal single-plex methods

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 2024 Thebiotek. All Rights Reserved.