At1g35537 Antibody

Shipped with Ice Packs
In Stock

Description

Current Status of At1g35537 Research

No peer-reviewed studies, commercial antibody catalogs, or structural databases (e.g., UniProt, PDB, or SabDab) describe an antibody targeting At1g35537. Key findings from the reviewed sources include:

  • Antibody specificity: All validated antibodies in the search results target human, viral, or mammalian proteins (e.g., SIRT1 , HIV CD4-binding site , influenza neuraminidase ).

  • Plant protein antibodies: While monoclonal antibodies are used in plant research (e.g., detecting pathogen antigens ), no examples target Arabidopsis gene products with identifier "At1g35537".

Technical Challenges in Developing At1g35537 Antibodies

The absence of this antibody aligns with broader trends in plant proteomics:

FactorImpact on Antibody Development
Low commercial demandPlant research antibodies represent <5% of the global antibody market
Uncharacterized targetsAt1g35537 lacks functional annotation in TAIR (The Arabidopsis Information Resource)
Sequence homologyNo cross-reactive epitopes identified in human/mammalian systems

Alternative Approaches for Studying At1g35537

Until specific antibodies are developed, researchers may employ:

  1. CRISPR/Cas9 knockouts: Assess phenotypic changes in Arabidopsis mutants .

  2. Transcriptomics: Measure expression via RNA-seq or qPCR.

  3. Heterologous expression: Express the protein in E. coli or yeast for in vitro assays.

Recommendations for Future Work

To advance this field:

  • Submit antigen design requests to companies like Abcam or Thermo Fisher with the At1g35537 protein sequence.

  • Explore phage display libraries for custom antibody generation .

  • Utilize structural prediction tools (AlphaFold2) to identify potential epitopes.

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M Phosphate Buffered Saline (PBS), pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
At1g35537 antibody; F15O4Defensin-like protein 32 antibody
Target Names
At1g35537
Uniprot No.

Target Background

Database Links

KEGG: ath:AT1G35537

STRING: 3702.AT1G35537.1

UniGene: At.63180

Protein Families
DEFL family
Subcellular Location
Secreted.

Q&A

How can I validate the specificity of At1g35537 antibodies?

Validating antibody specificity is crucial for ensuring experimental reliability and preventing non-specific binding. For At1g35537 antibodies, specificity validation should involve testing against genetically modified samples through several approaches. CRISPR-Cas9 gene editing to create At1g35537 knockout cell lines provides the most definitive validation method by comparing antibody binding in wild-type versus knockout samples. The complete absence of signal in knockout samples confirms specificity. Alternative methods include siRNA knockdown of At1g35537, which should show proportional reduction in antibody signal corresponding to the knockdown efficiency. Documentation of validation should include western blot analysis showing band presence in control samples and absence/reduction in knockout or knockdown samples .

What controls should be included when validating At1g35537 antibodies?

Proper validation requires multiple controls to ensure result reliability. Essential controls include:

  • Positive controls: Wild-type samples with confirmed At1g35537 expression

  • Negative controls:

    • At1g35537 knockout/knockdown samples

    • No-primary-antibody controls to assess secondary antibody non-specific binding

    • Isotype controls to evaluate potential non-specific binding

  • Loading controls: When performing western blots, include housekeeping proteins (like actin) to normalize protein loading across samples

Immunofluorescence experiments should include DAPI nuclear staining and cytoskeletal markers (e.g., phalloidin for F-actin) to provide cellular context and facilitate interpretation of At1g35537 localization patterns .

How should I design experiments to study At1g35537 protein interactions?

Studying At1g35537 protein interactions requires careful experimental design to capture both strong and transient interactions. Begin with co-immunoprecipitation (co-IP) using anti-At1g35537 antibodies to pull down protein complexes, followed by mass spectrometry to identify binding partners. For structural analysis of these interactions, crystallography methods similar to those used in therapeutic antibody research can provide valuable insights into binding mechanisms .

To validate identified interactions, employ reciprocal co-IPs and proximity ligation assays (PLA) which can detect protein interactions in situ. Consider using crosslinking approaches for capturing transient interactions. For each experiment, include negative controls (IgG isotype control, interaction-null mutants) to distinguish specific from non-specific interactions. The experimental workflow should systematically move from identification to validation and finally to functional characterization of each interaction .

What strategies can address epitope masking issues with At1g35537 antibodies?

Epitope masking occurs when protein-protein interactions or conformational changes block antibody access to the target epitope. For At1g35537 antibodies, several strategies can mitigate this issue:

  • Epitope retrieval methods:

    • Heat-induced epitope retrieval (HIER): Test multiple buffers (citrate, EDTA, Tris) at pH ranges 6-9

    • Enzymatic retrieval: Consider pepsin, trypsin, or proteinase K treatment with optimized concentration and incubation times

  • Multiple antibody approach: Use antibodies targeting different At1g35537 epitopes to increase detection probability

  • Denaturing conditions: For western blots, optimize SDS concentration and reducing agent strength

  • Fixation optimization: Compare results with different fixatives (paraformaldehyde, methanol, acetone) as fixation chemistry can differentially affect epitope accessibility

For each approach, conduct systematic optimization experiments with appropriate controls to identify the most effective combination of conditions for your specific experimental system .

What is the optimal immunofluorescence protocol for At1g35537 antibodies?

Optimizing immunofluorescence for At1g35537 antibodies requires attention to fixation, permeabilization, blocking, and antibody concentration. Based on protocols used for similar antibodies:

  • Fixation: Begin with 4% paraformaldehyde for 15 minutes at room temperature

  • Permeabilization: Use 0.1% Triton X-100 for 10 minutes

  • Blocking: Apply 1% BSA in PBS for 1 hour at room temperature

  • Primary antibody: Test At1g35537 antibody concentrations between 1-5 μg/mL in 0.1% BSA solution with overnight incubation at 4°C

  • Secondary antibody: Use species-appropriate fluorophore-conjugated secondary antibody (e.g., Alexa Fluor 488) at 1:2,000 dilution for 45 minutes

  • Counterstaining: Apply DAPI for nuclear visualization and phalloidin for F-actin staining

  • Mounting: Use anti-fade mountant to prevent photobleaching

Compare results across multiple fixation and permeabilization conditions, as these can significantly affect antibody performance. Document specificity through parallel staining of At1g35537 knockout or knockdown samples as negative controls .

How can I develop a quantitative ELISA for At1g35537 protein detection?

Developing a quantitative ELISA for At1g35537 requires careful optimization of multiple parameters:

  • Plate coating: Determine optimal antigen concentration (if direct ELISA) or capture antibody concentration (if sandwich ELISA)

  • Blocking buffer optimization: Test BSA, casein, and commercial blocking buffers at varying concentrations

  • Antibody dilution series: Create standard curves of primary and secondary antibody dilutions to identify optimal concentrations

  • Sample preparation: Optimize protein extraction and purification methods

  • Standard curve generation: Use recombinant At1g35537 protein at known concentrations

  • Detection system: Compare colorimetric, fluorescent, and chemiluminescent detection systems for sensitivity and dynamic range

To ensure specificity, include controls such as wells without antigen, without primary antibody, and with irrelevant antibodies. Validate the assay by showing proportional signal reduction when measuring samples with known decreasing concentrations of At1g35537 protein or when comparing wild-type to knockdown samples .

How can I address non-specific binding with At1g35537 antibodies?

Non-specific binding is a common challenge that can compromise experimental results. For At1g35537 antibodies, implement these approaches:

  • Optimize blocking conditions:

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

    • Extend blocking time (2-3 hours or overnight)

    • Incorporate 0.1-0.3% Tween-20 in washing buffers

  • Antibody dilution optimization:

    • Perform titration experiments to find minimum effective concentration

    • Pre-absorb antibodies with knockout or knockdown cell lysates

  • Cross-adsorption:

    • If cross-reactivity with related proteins is suspected, cross-adsorb antibodies against those purified proteins

  • Sample preparation modifications:

    • For tissue samples, incorporate additional washing steps

    • Consider antigen retrieval modifications for fixed samples

  • Alternative detection methods:

    • If western blots show multiple bands, try native PAGE or immunoprecipitation

    • If immunofluorescence shows diffuse staining, optimize fixation methods

Document all optimization steps methodically. Perform side-by-side comparisons with different antibody lots if available, as lot-to-lot variation can contribute to non-specific binding issues .

What strategies help troubleshoot weak or absent At1g35537 antibody signals?

When At1g35537 antibody signals are weak or absent despite confirmed target expression, systematic troubleshooting is required:

  • Sample preparation:

    • Verify protein extraction efficiency

    • Check for protein degradation with fresh protease inhibitors

    • Test different lysis buffers that may better preserve epitopes

  • Antibody factors:

    • Confirm antibody viability with positive controls

    • Test increased antibody concentration

    • Verify antibody storage conditions

  • Detection system enhancement:

    • For western blots, try more sensitive ECL substrates

    • For immunofluorescence, use signal amplification systems (tyramide, quantum dots)

    • Extend exposure times while monitoring background

  • Epitope accessibility:

    • Test different antigen retrieval methods

    • Try multiple antibodies targeting different epitopes

    • Consider native versus denaturing conditions

  • Technical modifications:

    • For western blots, reduce membrane washing stringency

    • For immunohistochemistry, extend primary antibody incubation time

    • Optimize buffer pH conditions

Each modification should be tested individually with appropriate controls to isolate the effective variables .

How can At1g35537 antibodies be utilized in multiplexed immunoassays?

Multiplexed immunoassays allow simultaneous detection of multiple targets, providing valuable contextual data about At1g35537 and its interaction partners or regulatory proteins. Implement these approaches:

  • Multiplex immunofluorescence:

    • Use antibodies from different host species

    • Employ sequential labeling with antibody stripping between rounds

    • Implement spectrally distinct fluorophores with minimal overlap

    • Consider tyramide signal amplification for weak signals

  • Multiplex western blotting:

    • Use antibodies with distinct molecular weight targets

    • Apply fluorescent secondary antibodies with different emission spectra

    • Implement sequential detection with stripping and reprobing

  • Mass cytometry (CyTOF):

    • Label At1g35537 antibodies with rare earth metals

    • Combine with antibodies against regulatory proteins or markers

  • Single-cell multiplexed analysis:

    • Integrate with single-cell RNA-seq data

    • Correlate protein expression with transcriptional profiles

For each approach, validation is essential - confirm that antibody performance in multiplexed formats matches that in single-target applications. Perform serial dilutions to ensure signal linearity and exclude antibody cross-reactivity .

How can I adapt At1g35537 antibodies for live-cell imaging applications?

Adapting At1g35537 antibodies for live-cell imaging requires careful consideration of antibody modification, delivery, and performance monitoring:

  • Antibody modification strategies:

    • Direct conjugation to small, bright fluorophores (Alexa dyes, DyLight, Atto dyes)

    • Use Fab fragments to improve tissue penetration

    • Consider nanobody alternatives if available

  • Delivery methods:

    • Microinjection for precise delivery with minimal cellular disruption

    • Cell-penetrating peptide conjugation

    • Electroporation with optimized parameters

    • Bead-loading techniques for adherent cells

  • Live-cell compatibility:

    • Test antibody performance in physiological buffers

    • Optimize antibody concentration to minimize functional interference

    • Verify cell viability throughout imaging sessions

  • Functional validation:

    • Confirm that antibody binding doesn't alter At1g35537 function

    • Compare protein dynamics with alternative tagging methods (e.g., fluorescent protein fusions)

    • Perform photobleaching experiments to assess off-target effects

  • Image acquisition optimization:

    • Use minimal light exposure to reduce phototoxicity

    • Implement deconvolution or super-resolution techniques for improved spatial resolution

    • Consider light-sheet microscopy for reduced phototoxicity during long-term imaging

Document all validation steps thoroughly and include appropriate controls to distinguish specific from non-specific signals in the dynamic cellular environment .

What approaches can be used to improve At1g35537 antibody specificity through engineering?

Antibody engineering offers powerful approaches to enhance At1g35537 antibody specificity:

  • Affinity maturation:

    • Implement phage display with stringent selection conditions

    • Use directed evolution with error-prone PCR

    • Apply rational design based on structural information

  • Complementarity-determining region (CDR) modification:

    • Identify and mutate key residues involved in non-specific interactions

    • Introduce additional hydrogen bonding or salt bridges with unique At1g35537 epitopes

  • Framework optimization:

    • Humanize antibodies to reduce background in human samples

    • Stabilize frameworks to improve thermodynamic properties

  • Bispecific approaches:

    • Create bispecific antibodies targeting At1g35537 plus a second unique marker

    • Require dual binding for detection, dramatically increasing specificity

  • Fragment-based approaches:

    • Use single-chain variable fragments (scFv) for improved tissue penetration

    • Implement camelid-derived single-domain antibodies (nanobodies) for accessing hidden epitopes

Each engineering approach requires thorough validation against both positive and negative controls, particularly testing against closely related proteins that might cross-react with the original antibody .

How can At1g35537 antibodies be adapted for proximity-dependent labeling applications?

Proximity-dependent labeling techniques enable mapping of protein interaction networks in native cellular environments. Adapting At1g35537 antibodies for these applications involves:

  • Enzyme conjugation strategies:

    • APEX2 peroxidase conjugation for biotin-phenol labeling

    • BioID/TurboID ligase conjugation for proximity-dependent biotinylation

    • HRP conjugation for tyramide signal amplification

  • Conjugation chemistry optimization:

    • Site-specific conjugation to avoid interfering with antigen binding

    • Testing various linker lengths to optimize enzyme positioning

    • Verifying retention of both antibody specificity and enzyme activity

  • Validation experiments:

    • Confirm labeling radius with known interaction partners

    • Verify specificity with knockout/knockdown controls

    • Compare results with orthogonal interaction detection methods

  • Data analysis approaches:

    • Implement quantitative proteomics for labeled protein identification

    • Apply statistical filtering to discriminate true interactors from background

    • Compare enrichment across multiple biological replicates and controls

  • Technical optimizations:

    • Adjust substrate concentration and labeling time

    • Optimize cell fixation timing (pre- vs. post-labeling)

    • Test various lysis conditions to maximize recovery of labeled proteins

This approach can reveal previously unknown At1g35537 interaction partners and provide insights into its functions within specific subcellular compartments .

How can AI and machine learning improve At1g35537 antibody-based image analysis?

Artificial intelligence and machine learning offer transformative potential for analyzing At1g35537 antibody-based imaging data:

  • Deep learning for image segmentation:

    • Train convolutional neural networks (CNNs) to automatically identify At1g35537-positive structures

    • Implement instance segmentation for quantifying individual structures

    • Use transfer learning to adapt pre-trained networks for At1g35537-specific detection

  • Automated pattern recognition:

    • Develop algorithms to classify subcellular localization patterns

    • Identify colocalization with other cellular markers

    • Quantify changes in localization under different experimental conditions

  • Multi-dimensional data integration:

    • Correlate imaging data with transcriptomics or proteomics

    • Integrate temporal dimensions for dynamic process analysis

    • Develop predictive models for protein function based on localization patterns

  • Quantitative analysis automation:

    • Standardize intensity measurements across experiments

    • Implement automated quality control for image acquisition

    • Develop pipelines for high-content screening applications

  • Validation approaches:

    • Train algorithms using ground truth data from multiple experts

    • Implement cross-validation across independent datasets

    • Compare algorithmic performance against human expert analysis

These approaches can dramatically increase throughput, reduce bias, and extract quantitative information beyond human visual capability from At1g35537 antibody-based imaging data1.

What are the future prospects for using At1g35537 antibodies in structural biology research?

Emerging technologies are expanding the potential applications of At1g35537 antibodies in structural biology:

  • Cryo-electron microscopy applications:

    • Use antibodies as fiducial markers for alignment

    • Implement antibody labeling for subunit identification in complexes

    • Apply Fab fragments to stabilize flexible regions for high-resolution imaging

  • X-ray crystallography approaches:

    • Use antibodies to facilitate crystallization of challenging proteins

    • Apply crystallography to determine At1g35537-antibody complex structures

    • Leverage structural information to design improved antibodies

  • Single-particle analysis:

    • Employ antibodies to identify specific conformational states

    • Use antibody binding to trap transient intermediates

    • Apply insights from structure-function relationships to understand mechanistic details

  • Integrative structural biology:

    • Combine antibody-based techniques with complementary methods (SAXS, NMR)

    • Develop computational approaches to integrate multiple structural datasets

    • Apply molecular dynamics simulations to antibody-antigen complexes

  • In-cell structural analysis:

    • Implement in-cell cryo-electron tomography with antibody labeling

    • Develop proximity-based structural elucidation techniques

    • Apply correlative light and electron microscopy with antibody labeling

These advanced approaches can provide unprecedented insights into At1g35537 structure, interactions, and conformational dynamics within its native cellular context .

What reporting standards should be followed when publishing research using At1g35537 antibodies?

Comprehensive reporting is essential for experimental reproducibility and scientific rigor when using At1g35537 antibodies:

  • Antibody identification:

    • Provide complete antibody information (supplier, catalog number, lot number, RRID)

    • Describe antibody type (monoclonal/polyclonal, host species, isotype)

    • Specify the immunogen used for antibody generation

  • Validation documentation:

    • Detail all validation experiments performed (western blot, immunofluorescence)

    • Include images of positive and negative controls

    • Describe knockdown/knockout validation if performed

  • Experimental protocols:

    • Provide complete methodology including buffer compositions

    • Specify antibody concentrations and incubation conditions

    • Detail sample preparation methods, including fixation parameters

  • Image acquisition and analysis:

    • Document microscope settings (exposure, gain, laser power)

    • Describe image processing methods in detail

    • Explain quantification approaches with statistical analyses

  • Data availability:

    • Deposit unprocessed images in appropriate repositories

    • Make analysis code publicly available

    • Consider antibody sharing to facilitate reproducibility

Following these reporting standards ensures that At1g35537 antibody-based research can be properly evaluated and reproduced by the scientific community .

How will advances in alternative protein detection technologies impact At1g35537 antibody use in research?

The landscape of protein detection technologies continues to evolve, offering both challenges and opportunities for At1g35537 antibody applications:

  • Mass spectrometry advancements:

    • Targeted proteomics approaches for absolute quantification

    • Improvements in sensitivity for low-abundance protein detection

    • Single-cell proteomics for heterogeneity analysis

  • Aptamer-based technologies:

    • Development of highly specific DNA/RNA aptamers as antibody alternatives

    • Aptamer-based proximity assays for protein interaction studies

    • Advantages in reproducibility and synthetic production

  • Genetic tagging methods:

    • CRISPR knock-in approaches for endogenous tagging

    • Split fluorescent protein complementation for interaction studies

    • Considerations of tag interference with protein function

  • Complementary roles:

    • Integration of antibody-based detection with orthogonal methods

    • Validation across multiple detection platforms

    • Selection of optimal technology based on specific research questions

  • Future directions:

    • Development of hybrid approaches combining antibodies with alternative technologies

    • Implementation of multiplexed detection systems

    • Standardization efforts across detection platforms

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.