At1g49180 Antibody

Shipped with Ice Packs
In Stock

Description

Molecular Characterization of At1g49180 (ATG1a)

Target Protein Structure:

  • Gene Locus: At1g49180 (Chromosome 1)

  • Protein: Autophagy-related 1a (ATG1a), part of the ATG1 kinase complex .

  • Domains:

    • N-terminal kinase domain

    • C-terminal regulatory domain interacting with ATG13 .

  • Molecular Weight: ~56.3 kDa (predicted) .

Antibody Specificity:

  • Validated for detecting ATG1a in immunoblotting (e.g., GFP-tagged ATG1a in transgenic lines) .

  • Cross-reactivity with other ATG1 isoforms (e.g., ATG1b, ATG1d) is minimal due to sequence divergence in variable regions .

Functional Role in Autophagy

ATG1a is a core component of the ATG1-ATG13 kinase complex, which initiates autophagosome formation during nutrient deprivation. Key findings include:

Autophagic Regulation Under Fixed-Carbon Starvation

ConditionATG1a ExpressionAutophagy Flux
Normal carbon supplyLowBaseline
Prolonged darknessUpregulatedIncreased
atg1a mutantsAbsentImpaired
  • ATG1a phosphorylation status dictates complex assembly with ATG13a .

  • ATG1a-deficient plants show delayed autophagosome formation and hypersensitivity to carbon starvation .

Comparative Analysis of ATG1 Isoforms

FeatureATG1a (At1g49180)ATG1bATG1d
Expression under stressHighModerateLow
Interaction with ATG13StrongWeakNone
Phenotype in mutantsLethal under stressViableViable

ATG1a is indispensable for autophagy induction, unlike its paralogs .

Key Studies Utilizing At1g49180 Antibody:

  1. Autophagosome Dynamics:

    • Immunoprecipitation (Co-IP) confirmed ATG1a-ATG13a interaction in response to carbon starvation .

    • Subcellular localization via fluorescence microscopy revealed punctate structures under stress .

  2. Stress Response Profiling:

    • Salt/Drought Stress: ATG1a levels correlate with autophagy activation in sweet orange (CsATG1a homolog) .

    • Pathogen Resistance: ATG1a-deficient plants exhibit compromised hypersensitive responses .

Technical Considerations

  • Host Species: Polyclonal antibodies are typically raised in rabbits for higher affinity .

  • Validation:

    • Western blotting using atg1a knockout controls .

    • GFP-ATG1a fusion constructs for in vivo tracking .

Challenges and Future Directions

  • Functional Redundancy: Despite ATG1a’s dominance, partial compensation by ATG1b/d complicates phenotypic analysis .

  • Post-Translational Modifications: Phosphorylation/dephosphorylation by TOPP phosphatases fine-tunes ATG1a activity .

Product Specs

Buffer
**Preservative:** 0.03% Proclin 300
**Constituents:** 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
At1g49180 antibody; F27J15.5Serine/threonine-protein kinase ATG1t antibody; EC 2.7.11.- antibody; Autophagy-related protein 1t antibody
Target Names
At1g49180
Uniprot No.

Target Background

Function
Serine/threonine protein kinase involved in autophagy. The ATG1-ATG13 protein kinase complex regulates downstream events required for autophagosome enclosure and/or vacuolar delivery.
Database Links

KEGG: ath:AT1G49180

STRING: 3702.AT1G49180.1

UniGene: At.38286

Protein Families
Protein kinase superfamily, Ser/Thr protein kinase family
Subcellular Location
Cytoplasmic vesicle, autophagosome.

Q&A

What is At1g49180 and why develop antibodies against it?

At1g49180 is a gene locus in Arabidopsis thaliana that encodes a specific protein. Developing antibodies against this protein enables researchers to study its expression patterns, localization, interactions, and functions within plant cells and tissues. Antibodies serve as crucial tools for protein detection through various techniques including Western blotting, immunoprecipitation, immunofluorescence, and ELISA. The development of specific antibodies against At1g49180 provides researchers with the capacity to investigate its biological roles in plant development, stress responses, or other physiological processes .

What are the different types of antibodies that can be developed against At1g49180?

Researchers can develop two main types of antibodies against At1g49180:

  • Polyclonal antibodies: Generated by immunizing animals with At1g49180 protein or peptide fragments, resulting in a heterogeneous mixture of antibodies that recognize different epitopes of the target protein. These are relatively faster and less expensive to produce but may have higher background and cross-reactivity issues.

  • Monoclonal antibodies: Produced using hybridoma technology where B lymphocytes from immunized animals are fused with myeloma cells to create immortalized cell lines that secrete antibodies with identical specificity for a single epitope. These offer higher specificity, reproducibility, and consistency but require more sophisticated development processes .

Each approach has distinct advantages depending on the research application, with monoclonal antibodies typically providing higher specificity for detailed protein characterization studies.

How is antibody specificity for At1g49180 validated?

Validating antibody specificity for At1g49180 requires multiple complementary approaches:

  • ELISA-based validation: Testing the antibody's ability to specifically recognize purified At1g49180 protein versus other control proteins .

  • Western blot analysis: Confirming the antibody detects a single band of the expected molecular weight in plant extracts expressing At1g49180 while showing minimal cross-reactivity with other proteins .

  • Immunoprecipitation followed by mass spectrometry: Verifying that the antibody enriches the target protein from complex biological samples.

  • Testing in knockout/knockdown lines: Demonstrating reduced or absent signal in plant lines where At1g49180 expression is eliminated or reduced.

  • Preabsorption tests: Pre-incubating the antibody with purified antigen to confirm that this eliminates specific staining in subsequent experiments .

These validation steps are essential to ensure experimental results accurately reflect At1g49180 biology rather than artifacts from cross-reactivity.

What immunization strategies work best for developing At1g49180 antibodies?

Effective immunization strategies for At1g49180 antibody development include:

  • Peptide-based immunization: Using synthetic peptides corresponding to unique, exposed regions of At1g49180, particularly those with high antigenicity and surface accessibility. This approach is advantageous when working with membrane proteins or when specific domains need targeting.

  • Recombinant protein immunization: Expressing and purifying the full At1g49180 protein or substantial domains for immunization, which may generate antibodies against multiple epitopes for better detection.

  • DNA immunization: Introducing DNA encoding At1g49180 directly into animal tissues, resulting in in vivo protein expression and immune response.

  • Adjuvant selection: Combining the antigen with appropriate adjuvants such as Freund's complete/incomplete adjuvant to enhance immune response .

  • Immunization schedule: Implementing proper timing between primary immunization and booster doses (typically 2-4 week intervals) to maximize antibody titer and affinity .

The selection of appropriate animal models (typically rabbits for polyclonal antibodies or mice for monoclonal antibodies) also significantly impacts antibody production success.

How can monoclonal antibody technology be optimized for At1g49180 detection?

Optimizing monoclonal antibody technology for At1g49180 detection requires several sophisticated approaches:

  • Epitope mapping and selection: Conducting computational analyses to identify highly antigenic, conserved, and accessible regions specific to At1g49180 to maximize antibody specificity and minimize cross-reactivity with related proteins.

  • Hybridoma screening optimization: Implementing high-throughput screening methods to efficiently identify hybridoma clones producing antibodies with optimal specificity and affinity for At1g49180.

  • Antibody production scale-up: After identifying promising hybridoma clones, researchers can either:

    • Culture hybridomas in bioreactors for supernatant collection

    • Produce antibodies in mouse ascites for higher yield (3 × 10^-5 mol/L from 1 × 10^7 hybridomas compared to 3.75 × 10^-6 mol/L from cell culture supernatants) .

  • Antibody engineering: Applying techniques like complementarity-determining region (CDR) modifications to improve binding affinity, as demonstrated in recent antibody design studies .

  • Validation in multiple plant tissues and developmental stages: Testing antibody performance across various experimental conditions to ensure consistent At1g49180 detection regardless of tissue type or developmental context.

This systematic approach ensures the development of high-quality monoclonal antibodies with optimal specificity and sensitivity for At1g49180 detection.

What are the challenges in distinguishing At1g49180 from closely related proteins?

Distinguishing At1g49180 from closely related proteins presents several technical challenges:

  • Sequence homology issues: Plant genomes often contain multiple related genes from the same family with high sequence similarity, requiring careful epitope selection to target unique regions of At1g49180.

  • Cross-reactivity assessment: Comprehensive testing against related proteins is essential, ideally using:

    • Recombinant related proteins in direct binding assays

    • Tissue samples from plants with knockouts of At1g49180 as negative controls

    • Competitive binding assays with purified related proteins

  • Splice variant considerations: At1g49180 may produce multiple splice variants, necessitating antibodies that either recognize common regions or specifically distinguish between variants.

  • Post-translational modifications: These modifications can affect epitope accessibility and antibody binding, requiring characterization of the protein's natural modification state.

  • Western blot optimization: Adjusting conditions such as blocking reagents, antibody concentration, and washing stringency to eliminate non-specific binding to related proteins .

Addressing these challenges requires iterative optimization and comprehensive validation across multiple experimental platforms.

How can At1g49180 antibodies be used to study protein-protein interactions?

At1g49180 antibodies can be powerful tools for studying protein-protein interactions through several advanced techniques:

  • Co-immunoprecipitation (Co-IP): Using At1g49180 antibodies to isolate the protein along with its binding partners from plant extracts, followed by mass spectrometry identification. This approach requires antibodies with high affinity for native protein conformations.

  • Proximity labeling methods: Combining At1g49180 antibodies with enzyme-conjugated secondary antibodies that generate reactive biotin species to label proximal proteins, enabling identification of transient or weak interactions.

  • Chromatin immunoprecipitation (ChIP): Applying At1g49180 antibodies to study protein-DNA interactions if the protein functions in transcriptional regulation or chromatin modification.

  • Förster resonance energy transfer (FRET): Using fluorophore-conjugated At1g49180 antibodies in combination with antibodies against potential interaction partners to detect molecular proximity in situ.

  • Bimolecular fluorescence complementation (BiFC): While not directly using antibodies, results from BiFC can be validated using At1g49180 antibodies to confirm protein expression and localization.

These methodologies provide complementary approaches to map the interactome of At1g49180, revealing its functional network within plant cellular processes.

What bioinformatic approaches aid in At1g49180 antibody design?

Advanced bioinformatic approaches significantly enhance At1g49180 antibody design:

  • Epitope prediction algorithms: Computational tools can identify regions of At1g49180 with high antigenicity, surface accessibility, and uniqueness compared to other proteome components.

  • Protein structure prediction: Using AlphaFold or similar platforms to predict the three-dimensional structure of At1g49180, identifying exposed regions optimal for antibody targeting.

  • Sequence alignment analysis: Comparing At1g49180 sequences across plant species to identify conserved regions for cross-species reactivity or variable regions for species-specific detection.

  • Machine learning-based antibody design: Novel approaches like DyAb that predict binding affinities for antibody variants can optimize binding properties through targeted mutations in complementarity-determining regions (CDRs) .

  • Post-translational modification prediction: Identifying potential modification sites that might interfere with antibody binding or create neo-epitopes.

These computational approaches can significantly reduce experimental trial-and-error, accelerating the development of high-performance antibodies against At1g49180.

What is the optimal protocol for purifying At1g49180 antibodies from hybridoma cultures?

The optimal protocol for purifying At1g49180 antibodies from hybridoma cultures involves several key steps:

  • Initial hybridoma selection and expansion:

    • Screen hybridoma clones for antibody production using ELISA against the At1g49180 antigen

    • Expand positive clones in appropriate media supplemented with fetal bovine serum

  • Antibody production methods:

    • In vitro culture: Grow hybridomas in suitable media for 3-5 days until reaching optimal density

    • In vivo production: Inject 1 × 10^7 hybridomas into mouse peritoneal cavity for ascites production (yields substantially higher antibody concentrations)

  • Purification procedure:

    • Centrifuge culture supernatant or ascites fluid to remove cells and debris

    • Perform ammonium sulfate precipitation to concentrate antibodies

    • Use affinity chromatography with Protein A/G columns for IgG purification

    • Consider additional purification steps (ion exchange chromatography) for highest purity

  • Quality control assessments:

    • Verify purity by SDS-PAGE (expect bands at approximately 55 kDa for heavy chains and 25 kDa for light chains)

    • Confirm specificity using Western blotting against plant extracts expressing At1g49180

    • Determine antibody concentration using BCA or similar protein assays

  • Storage conditions:

    • Store purified antibodies at -20°C or -80°C in small aliquots to avoid freeze-thaw cycles

    • Add appropriate preservatives (sodium azide 0.02%) for short-term storage at 4°C

This protocol typically yields antibody concentrations of 3 × 10^-5 mol/L from ascites compared to 3.75 × 10^-6 mol/L from cell culture supernatants, representing nearly an order of magnitude improvement in yield .

How should researchers optimize Western blot conditions for At1g49180 detection?

Optimizing Western blot conditions for At1g49180 detection requires systematic adjustment of multiple parameters:

  • Sample preparation optimization:

    • Select appropriate plant tissue extraction buffers containing protease inhibitors

    • Optimize protein extraction conditions (detergent types/concentrations for membrane proteins)

    • Determine optimal protein loading amount (typically 10-50 μg per lane)

  • Gel electrophoresis parameters:

    • Select appropriate acrylamide percentage based on At1g49180's molecular weight

    • Optimize running conditions to ensure clear band separation

    • Include molecular weight markers that span the expected size range

  • Transfer conditions:

    • Determine optimal transfer time and voltage for At1g49180's molecular weight

    • Select appropriate membrane type (PVDF often provides better sensitivity than nitrocellulose)

    • Verify transfer efficiency using reversible protein stains

  • Blocking and antibody incubation:

    • Test different blocking agents (BSA, milk, commercial blocking buffers)

    • Titrate primary antibody concentration (typically starting at 1:1000 and adjusting)

    • Optimize incubation times and temperatures for both primary and secondary antibodies

    • Include extensive washing steps with optimized buffer compositions

  • Detection system selection:

    • Choose between chemiluminescence, fluorescence, or colorimetric detection based on sensitivity requirements

    • Optimize exposure times for imaging

Each parameter should be systematically tested and optimized to achieve the highest signal-to-noise ratio for At1g49180 detection while minimizing background and non-specific binding.

What immunohistochemistry protocols work best for localizing At1g49180 in plant tissues?

Effective immunohistochemistry protocols for localizing At1g49180 in plant tissues include:

  • Tissue fixation and embedding:

    • Fix plant tissues in 4% paraformaldehyde or other aldehyde-based fixatives

    • Consider the nature of At1g49180 (membrane-bound, soluble, etc.) when selecting fixation methods

    • Embed in paraffin for thin sectioning or prepare cryosections for better epitope preservation

  • Antigen retrieval techniques:

    • Apply heat-induced epitope retrieval using citrate buffer (pH 6.0)

    • Test enzymatic antigen retrieval if heat-based methods are insufficient

    • Optimize retrieval conditions to balance epitope exposure with tissue morphology preservation

  • Blocking and antibody application:

    • Block with BSA (3-5%) containing normal serum from the secondary antibody species

    • Apply optimized dilution of At1g49180 primary antibody (determined through titration)

    • Include negative controls (primary antibody omission, pre-immune serum, competing peptide)

  • Detection systems:

    • Use fluorescent secondary antibodies for co-localization studies

    • Apply peroxidase/alkaline phosphatase systems for bright-field microscopy

    • Consider tyramide signal amplification for low-abundance proteins

  • Counterstaining and mounting:

    • Apply appropriate counterstains to visualize cellular structures

    • Use mounting media with anti-fade properties for fluorescence preservation

This methodology should be optimized for each specific plant tissue type, with particular attention to fixation conditions that preserve both tissue architecture and At1g49180 antigenicity.

How can ELISA be optimized for quantitative analysis of At1g49180 protein levels?

Optimizing ELISA for quantitative analysis of At1g49180 protein levels requires careful attention to several critical factors:

  • ELISA format selection:

    • Direct ELISA: Simplest format but may have higher background

    • Sandwich ELISA: Higher specificity using two antibodies recognizing different epitopes

    • Competitive ELISA: Useful when only one antibody is available or for small proteins

  • Protocol optimization steps:

    • Coating concentration: Determine optimal concentration of capture antibody or antigen

    • Blocking conditions: Test different blocking agents (BSA, milk proteins, commercial blockers)

    • Sample preparation: Develop standardized extraction procedures for plant tissues

    • Antibody dilutions: Establish optimal primary and secondary antibody concentrations

    • Incubation conditions: Optimize times and temperatures for all steps

  • Standard curve development:

    • Prepare recombinant At1g49180 protein as a quantitative standard

    • Create a dilution series covering the expected concentration range

    • Ensure the standard curve encompasses the linear detection range

  • Signal detection optimization:

    • Select appropriate enzyme/substrate combination (HRP/TMB, AP/pNPP)

    • Determine optimal development time for maximum sensitivity

    • Measure absorbance at the appropriate wavelength (typically 405-450 nm)

  • Data analysis and validation:

    • Apply appropriate curve-fitting models for standard curves

    • Calculate protein concentrations from optical density readings

    • Validate results with spike-recovery experiments and precision assessment

The optimized ELISA should have a detection limit appropriate for the biological range of At1g49180 in plant samples with sufficient sensitivity and specificity for reliable quantification.

How can researchers address non-specific binding issues with At1g49180 antibodies?

Researchers can address non-specific binding issues with At1g49180 antibodies through several targeted approaches:

  • Antibody purification enhancements:

    • Implement affinity purification against the specific immunizing antigen

    • Consider subtractive purification against related plant proteins

    • Use protein A/G purification to isolate IgG fractions only

  • Blocking optimization:

    • Test alternative blocking agents (BSA, casein, commercial blockers)

    • Increase blocking agent concentration or incubation time

    • Add low concentrations of detergents (0.05-0.1% Tween-20) to reduce hydrophobic interactions

  • Buffer optimization:

    • Adjust salt concentration in washing and incubation buffers (150-500 mM NaCl)

    • Modify buffer pH to reduce non-specific interactions

    • Add competing agents like non-fat dry milk or non-related plant extracts

  • Antibody incubation adjustments:

    • Reduce primary antibody concentration

    • Shorten incubation times or adjust temperature

    • Pre-absorb antibodies with plant extracts lacking At1g49180

  • Additional controls and validation:

    • Perform peptide competition assays to confirm specificity

    • Use knockout/knockdown plant lines as negative controls

    • Conduct Western blots alongside immunoassays to confirm specificity

These approaches should be tested systematically, ideally changing one variable at a time to identify the optimal conditions for reducing non-specific binding while maintaining strong specific signal for At1g49180.

What controls are essential when using At1g49180 antibodies in experimental systems?

Essential controls when using At1g49180 antibodies include:

  • Positive controls:

    • Purified recombinant At1g49180 protein or peptide

    • Plant tissues known to express At1g49180 at high levels

    • Transgenic plants overexpressing At1g49180

  • Negative controls:

    • Knockout/knockdown plants lacking At1g49180 expression

    • Pre-immune serum from the same animal used to generate antibodies

    • Primary antibody omission control

    • Isotype control antibodies (particularly for monoclonal antibodies)

  • Specificity controls:

    • Peptide competition/blocking experiments where antibody is pre-incubated with immunizing antigen

    • Western blot analysis showing a single band of expected molecular weight

    • Immunoprecipitation followed by mass spectrometry identification

  • Technical controls:

    • Loading controls for Western blots (housekeeping proteins)

    • Staining controls for immunohistochemistry (counterstains for tissue morphology)

    • Standard curves for quantitative applications

  • Cross-reactivity assessment:

    • Testing antibody against related proteins or plant species

    • Gradient dilution series to assess signal specificity thresholds

Implementation of these controls ensures experimental reliability and supports valid interpretation of results when studying At1g49180 biology.

How should researchers interpret conflicting results from different At1g49180 antibody-based assays?

When faced with conflicting results from different At1g49180 antibody-based assays, researchers should follow this systematic approach:

  • Antibody characterization analysis:

    • Compare epitopes recognized by different antibodies (N-terminal, C-terminal, internal domains)

    • Assess antibody class and subclass (polyclonal vs. monoclonal, IgG subtype)

    • Review validation data for each antibody (Western blot, immunoprecipitation, ELISA results)

  • Technical considerations:

    • Evaluate whether differences arise from assay conditions rather than antibody specificity

    • Determine if protein denaturation affects epitope accessibility differently between assays

    • Consider whether post-translational modifications might affect antibody binding in different contexts

  • Biological variables assessment:

    • Analyze whether conflicting results correspond to different At1g49180 splice variants

    • Investigate developmental or stress-induced changes that might alter protein conformation

    • Examine whether protein-protein interactions could mask epitopes in certain assays

  • Resolution strategies:

    • Perform parallel experiments using multiple antibodies against different epitopes

    • Complement antibody-based approaches with orthogonal methods (mass spectrometry, RNA analysis)

    • Develop knockdown or knockout controls to validate all antibodies simultaneously

  • Data integration framework:

    • Create a coherent model that explains observed discrepancies

    • Weight evidence based on control validation strength

    • Consider biological plausibility when interpreting conflicting results

This structured approach helps researchers distinguish genuine biological phenomena from technical artifacts when working with At1g49180 antibodies.

What are the best practices for long-term storage and handling of At1g49180 antibodies?

Best practices for long-term storage and handling of At1g49180 antibodies include:

  • Initial processing and aliquoting:

    • Purify antibodies to highest possible purity before storage

    • Divide into small working aliquots (20-50 μL) to minimize freeze-thaw cycles

    • Use sterile, low-protein binding microcentrifuge tubes

  • Storage buffer optimization:

    • Store in phosphate or Tris buffer (pH 7.2-7.6) with 150 mM NaCl

    • Add protein stabilizers such as BSA (1-5 mg/mL) or gelatin (0.1%)

    • Include sodium azide (0.02-0.05%) as preservative for 4°C storage

    • Consider adding glycerol (30-50%) for freezer storage

  • Temperature conditions:

    • Long-term storage: -80°C preferred (maintains activity for years)

    • Medium-term storage: -20°C acceptable (stable for months)

    • Working aliquots: 4°C with preservative (stable for weeks)

    • Avoid repeated freeze-thaw cycles (ideally <5 total)

  • Handling procedures:

    • Allow antibodies to warm to room temperature before opening tubes

    • Centrifuge briefly before opening to collect condensation

    • Use clean, sterile pipette tips for each access

    • Return to appropriate storage temperature promptly after use

  • Quality monitoring:

    • Record date of preparation and number of freeze-thaw cycles

    • Periodically test activity against positive controls

    • Monitor for signs of degradation (precipitation, loss of activity)

    • Consider adding protease inhibitors for additional protection

Following these guidelines will maximize antibody stability and ensure consistent experimental results over extended research periods.

How are current advances in antibody engineering applicable to At1g49180 research?

Recent advances in antibody engineering offer significant opportunities for enhancing At1g49180 research:

  • Targeted affinity enhancement: New computational approaches like DyAb allow for sequence-based antibody design that can predict and improve binding affinity through strategic amino acid substitutions in complementarity-determining regions (CDRs) . These methods could be applied to existing At1g49180 antibodies to enhance their sensitivity and specificity.

  • Genetic engineering platforms: CRISPR-based antibody engineering enables precise modification of antibody genes to improve properties such as stability, specificity, and affinity, potentially creating superior At1g49180 detection reagents.

  • Fragment-based approaches: Generating single-chain variable fragments (scFvs) or antigen-binding fragments (Fabs) against At1g49180 could improve tissue penetration and reduce background in imaging applications by eliminating the Fc region .

  • Bifunctional antibody development: Creating bispecific antibodies that simultaneously recognize At1g49180 and another protein of interest could enable novel co-localization or proximity-based studies of protein-protein interactions.

  • Recombinant expression systems: Advanced expression platforms allow for the production of fully human or humanized antibodies with reduced immunogenicity for therapeutic applications, while plant-based expression systems might be particularly suitable for generating antibodies against plant proteins like At1g49180.

These technological advances promise to expand the toolkit available for At1g49180 research, enabling increasingly sophisticated studies of its biological functions and interactions.

What emerging techniques complement antibody-based detection of At1g49180?

Several emerging techniques complement traditional antibody-based approaches for At1g49180 detection and characterization:

  • CRISPR-based tagging: Direct genomic tagging of At1g49180 with fluorescent proteins or epitope tags using CRISPR-Cas9 enables visualization and purification without requiring specific antibodies.

  • Proximity labeling approaches: Methods like BioID or TurboID can be used to fuse promiscuous biotin ligases to At1g49180, enabling identification of proximal interacting proteins without requiring antibodies for each potential interactor.

  • Nanobody technology: Single-domain antibodies derived from camelids offer smaller size, enhanced stability, and the ability to recognize epitopes inaccessible to conventional antibodies, potentially improving At1g49180 detection in complex samples.

  • Mass spectrometry advances: Targeted proteomics approaches like parallel reaction monitoring (PRM) or selected reaction monitoring (SRM) enable quantitative detection of At1g49180 peptides without antibodies.

  • DNA aptamer development: Systematic evolution of ligands by exponential enrichment (SELEX) can generate DNA aptamers specific to At1g49180 that function as "chemical antibodies" with potentially superior stability and production consistency.

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.