At1g60730 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Preservative: 0.03% Proclin 300; Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
14-16 week lead time (made-to-order)
Synonyms
At1g60730 antibody; F8A5.24Probable aldo-keto reductase 5 antibody; EC 1.1.1.- antibody
Target Names
At1g60730
Uniprot No.

Q&A

Basic Questions on Antibody Production

Q: What are the most effective approaches for generating antibodies against the At1g60730 protein?

The generation of high-quality antibodies against plant proteins like At1g60730 typically involves using recombinant proteins expressed in eukaryotic expression systems as immunogens. Evidence indicates that using eukaryotic expression systems rather than bacterial systems is particularly important for heavily glycosylated plant proteins, as this better preserves native protein conformations and post-translational modifications . The process generally involves protein purification, immunization of animals (typically mice for monoclonal or rabbits for polyclonal antibodies), followed by hybridoma development or serum collection, respectively.

For proteins like At1g60730, which may contain complex structural elements, immunizing with carefully selected antigenic regions rather than the full-length protein often yields antibodies with superior specificity. Researchers should consider both N-terminal and C-terminal regions as potential immunogens, evaluating which domains are most accessible in the native protein conformation and least conserved among related proteins.

Fusion partners play a critical role in successful hybridoma development, with spleen cells from immunized animals typically fused with myeloma cell lines such as SP2/0-Ag14 to generate stable antibody-producing cell lines . This technique is essential for developing reproducible monoclonal antibodies with consistent binding properties.

Q: What are the comparative advantages of monoclonal versus polyclonal antibodies for At1g60730 detection?

Monoclonal antibodies offer superior consistency and specificity for At1g60730 detection compared to polyclonal alternatives. Since monoclonal antibodies derive from a single B-cell clone, they recognize a single epitope, minimizing cross-reactivity with related plant proteins. This characteristic is particularly valuable in Arabidopsis research where protein families often contain closely related members with high sequence homology.

Polyclonal antibodies, while easier to produce, introduce significant reproducibility challenges due to their heterogeneous nature. As highlighted in recent reviews, polyclonal antibodies represent "a large source of problems" in research due to "their non-renewable nature and the complexity of different antibodies present, which can influence batch variability" . This variability is particularly problematic in longitudinal studies where consistent antibody performance is essential.

Monoclonal antibodies typically require only "1 cell fusion and 2 cyclic sub-cloning steps" before obtaining antibodies with "satisfactory performance" , making them increasingly preferred for plant protein detection despite higher initial development costs. Their renewable nature ensures experimental consistency across different studies and laboratories.

Q: What basic validation steps are essential before using a new At1g60730 antibody?

Before implementing a new At1g60730 antibody in experimental workflows, researchers must conduct comprehensive validation procedures. At minimum, specificity should be demonstrated through Western blotting against both recombinant At1g60730 protein and native plant extracts. The antibody should detect a band of the predicted molecular weight with minimal cross-reactivity.

Immunoprecipitation followed by mass spectrometry provides a robust validation approach, confirming that the antibody specifically captures the intended target. Additionally, comparing antibody reactivity between wild-type and At1g60730 knockout or knockdown Arabidopsis lines represents a critical negative control. As emphasized in recent literature, "the use of knockout (KO) or knockdown (KD) cell lines or tissue samples as important negative controls for specificity" has become increasingly accessible due to CRISPR technologies .

Validation should also include immunohistochemistry or immunofluorescence experiments to confirm expected localization patterns of the At1g60730 protein. Cross-validation with multiple techniques strengthens confidence in antibody specificity before proceeding to experimental applications.

Advanced Questions on Specificity and Characterization

Q: How can researchers definitively characterize the epitope recognized by an At1g60730 antibody?

Epitope characterization for At1g60730 antibodies requires a systematic approach combining biochemical, structural, and computational methods. Precise epitope identification begins with epitope mapping using overlapping peptide arrays or phage display libraries expressing protein fragments. For instance, approaches similar to those used with plant cell wall antibodies can be adapted, where researchers precisely defined epitopes as specific as "trimer of beta-(1,6)-Gal, some substitution of single Ara or beta-(1,3)-Gal is tolerated, but not at 3- or 4-position of the central 6-Gal" .

Advanced epitope characterization involves competitive binding assays, where synthetic peptides corresponding to predicted epitope regions are tested for their ability to block antibody binding. Decreasing signal intensity in the presence of specific peptides identifies the epitope region. For conformational epitopes common in native plant proteins, hydrogen-deuterium exchange mass spectrometry (HDX-MS) can identify antibody-protein interaction surfaces by measuring changes in deuterium uptake when the antibody binds.

Computational approaches complement experimental data, with molecular docking and molecular dynamics simulations predicting epitope-paratope interactions. X-ray crystallography of antibody-antigen complexes, while technically challenging, provides definitive structural information about the binding interface. This comprehensive characterization informs interpretation of experimental results and guides antibody application optimization.

Q: What strategies can address cross-reactivity issues with At1g60730 antibodies in complex plant extracts?

Cross-reactivity represents a significant challenge when working with plant antibodies, particularly in complex tissue extracts containing numerous related proteins. For At1g60730 antibodies, researchers should implement multiple parallel strategies to minimize and characterize potential cross-reactivity.

Affinity purification represents a primary approach, where antibodies are purified using immobilized recombinant At1g60730 protein. This technique, described as "affinity chromatography on Protein A from tissue culture supernatant" for other antibodies, significantly reduces non-specific binding by isolating only those antibody molecules with high affinity for the target protein.

Pre-adsorption protocols, where antibodies are incubated with extracts from At1g60730 knockout plants before use, can effectively remove antibodies that bind non-specific targets. Complementary inhibition assays, where increasing concentrations of purified At1g60730 protein progressively reduce antibody binding to blots or cells, confirm binding specificity. Signal reduction correlates with epitope specificity.

For particularly challenging applications, researchers might consider developing bispecific antibodies that recognize two distinct epitopes on At1g60730, dramatically increasing specificity. This approach parallels advanced methodologies where researchers "engineered the nanobodies into a triple tandem format—by repeating short lengths of DNA—the resulting nanobodies demonstrated remarkable effectiveness" in other research contexts.

Basic Experimental Design Questions

Q: What are the optimal sample preparation methods for detecting At1g60730 in different plant tissues?

Effective detection of At1g60730 protein requires tissue-specific sample preparation protocols that preserve protein integrity while maximizing extraction efficiency. For Arabidopsis tissues, researchers should consider developmental stage, tissue type, and protein localization when selecting extraction methods.

For leaf tissue, grinding in liquid nitrogen followed by extraction in a buffer containing 50mM Tris-HCl (pH 7.5), 150mM NaCl, 1% Triton X-100, 0.5% sodium deoxycholate, and protease inhibitor cocktail typically yields good results. For membrane-associated proteins, inclusion of 0.1% SDS may improve solubilization while maintaining antibody compatibility. Root tissues often contain higher levels of interfering compounds, necessitating additional purification steps such as TCA/acetone precipitation prior to analysis.

Sample preparation for immunohistochemistry requires particular attention to fixation conditions. Paraformaldehyde (4%) fixation for 4 hours followed by paraffin embedding preserves antigenicity while maintaining tissue architecture. For challenging epitopes, researchers should test alternative fixatives or implement antigen retrieval techniques, such as heat-induced epitope retrieval in citrate buffer (pH 6.0).

For all applications, researchers should include multiple biological replicates and appropriate controls. Extracts from At1g60730 knockout plants provide the gold standard negative control, while extracts from plants overexpressing At1g60730 serve as positive controls, enabling accurate interpretation of signal specificity and sensitivity.

Q: What western blotting conditions work best for At1g60730 antibody detection?

Optimizing western blotting conditions for At1g60730 detection requires systematic evaluation of multiple parameters to maximize signal-to-noise ratio. Based on technical protocols established for other plant antibodies, researchers should start with antibody dilutions between 1/500 and 1/1000 as a baseline for optimization . Primary antibody incubation should be performed overnight at 4°C in blocking buffer containing 5% non-fat dry milk in TBST (Tris-buffered saline with 0.1% Tween-20).

Protein transfer conditions significantly impact detection efficiency, with semi-dry transfer using PVDF membranes generally providing optimal results for plant proteins like At1g60730. Transfer should be performed at 15V for 30 minutes to ensure complete transfer of proteins up to 100 kDa while minimizing potential membrane overheating.

Signal development options include both chemiluminescence and fluorescence-based detection. Enhanced chemiluminescence offers high sensitivity for low-abundance proteins, while fluorescence-based detection provides superior quantitative accuracy and multiplexing capabilities. For particularly challenging detection scenarios with low signal strength, researchers might implement signal amplification systems like tyramide signal amplification or polymer-based detection.

The inclusion of technical controls is essential, particularly loading controls appropriate for plant tissues such as actin, tubulin, or GAPDH. Signal normalization against these housekeeping proteins enables accurate quantification and comparison across multiple samples and experimental conditions.

Q: How can flow cytometry be optimized for At1g60730 analysis in plant protoplasts?

Flow cytometric analysis of At1g60730 in plant protoplasts requires specialized protocols that address the unique characteristics of plant cells. Protoplast preparation represents the critical first step, with enzymatic digestion using 1.5% cellulase R10 and 0.4% macerozyme R10 in a solution containing 0.4M mannitol, 20mM KCl, and 20mM MES (pH 5.7) typically yielding viable protoplasts suitable for antibody staining.

Fixation with 2% paraformaldehyde for 15 minutes at room temperature, followed by permeabilization with 0.1% Triton X-100 for 10 minutes, enables antibody access to intracellular proteins while maintaining cellular integrity. Blocking with 3% BSA reduces non-specific binding. Following principles established for other antibodies, staining should employ primary antibody at concentrations between 1-5 μg/mL for 60 minutes, followed by appropriate fluorophore-conjugated secondary antibodies .

Flow cytometer settings require careful optimization, with forward and side scatter parameters adjusted to accommodate the larger size and different morphology of plant protoplasts compared to animal cells. Including viability dyes such as propidium iodide for non-permeabilized samples helps exclude dead cells from analysis.

Importantly, gating strategies should incorporate negative controls (protoplasts from At1g60730 knockout plants), isotype controls, and positive controls when available. As demonstrated with other antibodies, this approach enables reliable "detecting of PD-L1/B7-H1 in MDA-MB-231 human breast adenocarcinoma cell line by Flow Cytometry" , and similar rigorous controls should be applied to plant cell analysis.

Advanced Experimental Considerations

Q: What approaches can detect protein-protein interactions involving At1g60730 using antibody-based methods?

Investigating protein-protein interactions involving At1g60730 requires specialized antibody-based approaches that preserve native protein complexes. Co-immunoprecipitation (Co-IP) represents the foundation of such studies, wherein anti-At1g60730 antibodies capture both the target protein and its interacting partners from plant extracts. For optimal results, researchers should employ gentle lysis conditions (1% NP-40 or 0.5% digitonin) that maintain protein-protein interactions while achieving efficient extraction.

Proximity ligation assays (PLA) offer an advanced in situ approach for detecting At1g60730 interactions within intact plant tissues. This technique uses primary antibodies against At1g60730 and its suspected interaction partner, followed by species-specific secondary antibodies conjugated to complementary oligonucleotides. When proteins interact closely (<40 nm), these oligonucleotides can be ligated and amplified, generating a fluorescent signal that precisely localizes the interaction within cellular compartments.

For quantitative analysis of dynamic interactions, bioluminescence resonance energy transfer (BRET) or fluorescence resonance energy transfer (FRET) may be combined with antibody-based approaches. By fusing At1g60730 and potential interactors with appropriate fluorescent or luminescent tags, researchers can monitor interaction kinetics in response to developmental cues or environmental stimuli.

Cross-linking mass spectrometry represents a powerful companion technique, where proteins are chemically cross-linked in vivo before immunoprecipitation with At1g60730 antibodies. Subsequent proteomic analysis identifies cross-linked peptides, providing structural insights into interaction interfaces. This multi-faceted approach builds a comprehensive understanding of At1g60730's interaction network within plant cellular systems.

Q: How can ChIP-seq be optimized using At1g60730 antibodies to study protein-DNA interactions?

Chromatin immunoprecipitation followed by sequencing (ChIP-seq) using At1g60730 antibodies requires meticulous optimization to generate reliable genome-wide binding profiles. Initial validation should establish that the antibody efficiently immunoprecipitates At1g60730 under ChIP conditions, as formaldehyde fixation can alter epitope accessibility. Testing antibody performance in ChIP-qPCR experiments against known or predicted binding sites provides essential validation before proceeding to genome-wide analysis.

Cross-linking conditions represent a critical parameter requiring optimization. While standard protocols employ 1% formaldehyde for 10 minutes, proteins with indirect DNA interactions may require alternative cross-linkers such as disuccinimidyl glutarate (DSG) followed by formaldehyde, which better preserves protein-protein interactions within DNA-binding complexes.

Chromatin fragmentation significantly impacts ChIP-seq resolution and efficiency. Sonication should be optimized to generate fragments between 200-500 bp, with fragment size verified by agarose gel electrophoresis. For plant tissues, which often contain interfering compounds, additional purification steps may be necessary to improve antibody binding specificity.

Input normalization and appropriate controls are essential for accurate data interpretation. In addition to input chromatin, ChIP-seq experiments should include negative controls (IgG or pre-immune serum) and, ideally, samples from At1g60730 knockout plants. These controls enable identification of artifacts and non-specific binding events during computational analysis of sequencing data, improving the reliability of identified binding sites.

Basic Troubleshooting

Q: What are common causes of high background when using At1g60730 antibodies in immunofluorescence?

High background in immunofluorescence experiments with At1g60730 antibodies typically stems from multiple factors that require systematic troubleshooting. Non-specific binding to endogenous plant components represents a primary cause, particularly in Arabidopsis tissues rich in phenolic compounds and cell wall components that can adsorb antibodies.

Optimizing blocking conditions is essential for reducing non-specific binding. While standard protocols typically employ 3-5% BSA, plant tissues often benefit from alternative blocking agents such as 5% normal goat serum, 2% non-fat dry milk, or specialized commercial blocking solutions designed for plant samples. Additionally, adding 0.1-0.3% Triton X-100 to blocking and antibody incubation buffers improves penetration while reducing hydrophobic interactions.

Autofluorescence from chlorophyll, lignin, and other plant compounds frequently confounds immunofluorescence imaging. Pre-treatment with 0.1% sodium borohydride can reduce aldehyde-mediated autofluorescence, while Sudan Black B (0.1% in 70% ethanol) effectively quenches lipofuscin-like autofluorescence. Selecting fluorophores with emission spectra distinct from plant autofluorescence (far-red rather than green) also improves signal discrimination.

Secondary antibody cross-reactivity contributes significantly to background issues. Researchers should pre-adsorb secondary antibodies against plant tissue extracts from At1g60730 knockout plants and include secondary-only controls in all experiments. Employing directly conjugated primary antibodies eliminates secondary antibody-related background entirely, though at the cost of reduced signal amplification.

Q: How can researchers address inconsistent western blot results with At1g60730 antibodies?

Inconsistent western blot results with At1g60730 antibodies typically stem from variability in sample preparation, protein transfer efficiency, or antibody performance across experiments. Standardizing lysate preparation represents the first critical step toward consistency, with researchers implementing rigorous protocols for tissue harvesting, extraction buffer composition, and protein quantification. Including protease inhibitor cocktails is essential to prevent degradation of the target protein during extraction.

Transfer efficiency significantly impacts detection consistency, particularly for membrane-associated or highly glycosylated proteins. Staining membranes with Ponceau S after transfer provides visual confirmation of consistent transfer and loading, enabling normalization during subsequent analysis. For problematic proteins, researchers might consider alternative transfer methods, such as wet transfer or specialized buffer systems optimized for glycoproteins.

Antibody performance variability remains a significant challenge, particularly with polyclonal antibodies where "the profile of a polyclonal antibody response can vary over time, even with affinity purification, as the antibody population in each batch is varied" . To mitigate this issue, researchers should prepare large antibody aliquots stored at -80°C to minimize freeze-thaw cycles, as recommended for other antibodies: "Storage in frost-free freezers is not recommended" and researchers should "avoid repeated freezing and thawing as this may denature the antibody" .

Positive controls are invaluable for troubleshooting inconsistent results, with recombinant At1g60730 protein or extracts from plants overexpressing the protein serving as reliable benchmarks for antibody performance. Including these controls in every experiment enables researchers to distinguish between technical issues and biological variability.

Q: What strategies effectively enhance detection sensitivity for low-abundance At1g60730 protein?

Detecting low-abundance At1g60730 protein requires implementing sensitivity-enhancing strategies across multiple stages of the experimental workflow. Sample enrichment techniques represent the first approach, with immunoprecipitation using the At1g60730 antibody followed by western blotting (IP-western) concentrating the target protein from larger sample volumes. Subcellular fractionation similarly enriches samples by isolating the cellular compartment where At1g60730 predominantly localizes.

Signal amplification systems dramatically improve detection sensitivity for immunoblotting and immunohistochemistry. Tyramide signal amplification (TSA) can increase sensitivity by 10-100 fold by generating multiple fluorophore deposits at antibody binding sites. Polymer-based detection systems, which carry multiple enzyme molecules per antibody binding event, similarly enhance chromogenic or chemiluminescent signal development.

Advanced imaging technologies significantly improve detection of weak fluorescence signals in microscopy applications. Structured illumination microscopy (SIM) increases resolution while reducing background, while deconvolution algorithms enhance signal clarity by mathematically removing out-of-focus light. For extreme sensitivity, techniques like single-molecule localization microscopy can detect individual molecules of target protein within plant cells.

Modern highly-sensitive Western blotting substrates can detect femtogram quantities of protein. Similarly, utilizing high-efficiency blocking solutions containing both proteins and polymeric compounds can reduce background while preserving specific signals. These combined approaches enable detection of even trace amounts of At1g60730 protein in complex plant extracts.

Advanced Technical Considerations

Q: How can researchers detect post-translational modifications of At1g60730 using modified antibody approaches?

Detecting post-translational modifications (PTMs) of At1g60730 requires specialized antibody strategies that selectively recognize modified epitopes. Modification-specific antibodies represent the cornerstone of this approach, with antibodies raised against synthetic peptides containing the specific modification of interest (phosphorylation, ubiquitination, SUMOylation, etc.). These antibodies should undergo rigorous validation using positive controls (in vitro modified recombinant At1g60730) and negative controls (modification-deficient mutants).

For phosphorylation studies, researchers can implement differential Western blotting, where parallel samples are treated with phosphatase before analysis. Signal reduction after phosphatase treatment confirms phosphorylation-specific antibody binding. Similar approaches apply to other reversible modifications, with specific enzymes used to remove the modification of interest.

Mass spectrometry-based validation strengthens antibody-based PTM detection by providing site-specific modification information. Immunoprecipitation of At1g60730 using general antibodies followed by mass spectrometry analysis identifies modified residues, which can subsequently be targeted with site-specific modification antibodies for larger-scale studies.

Multiplexed detection systems enable simultaneous visualization of total At1g60730 and its modified forms, providing valuable information about modification stoichiometry. Two-color Western blotting using differentially labeled secondary antibodies, or sequential probing with total and modification-specific antibodies, enables direct comparison of modified versus unmodified protein populations under different experimental conditions.

Q: What are the most effective approaches for multiplex detection of At1g60730 alongside other proteins of interest?

Multiplex detection of At1g60730 alongside other proteins enables researchers to investigate complex biological relationships within the same sample. Implementing successful multiplexing requires careful antibody selection and detection system optimization to prevent cross-reactivity while maintaining sensitivity for each target.

Fluorescence-based Western blotting with spectrally distinct fluorophores enables simultaneous detection of multiple proteins on a single membrane. This approach requires primary antibodies from different host species paired with species-specific secondary antibodies conjugated to compatible fluorophores. Digital imaging systems with appropriate filter sets can then distinguish between signals from each target protein. This approach parallels established protocols where researchers demonstrated "binding is completely blocked by 2.5 μg/mL of Human Anti-Human PD-L1/B7-H1 (Research Grade Atezolizumab Biosimilar) Monoclonal Antibody" while simultaneously detecting other markers.

For immunohistochemistry applications, multiplexing can be achieved through sequential detection protocols, where antibody-antigen complexes from each round of staining are either chemically stripped or permanently labeled before proceeding to the next target. Tyramide signal amplification systems are particularly valuable in this context, as they enable covalent deposition of fluorophores that withstand subsequent antibody stripping steps.

Mass cytometry (CyTOF) represents an advanced approach for highly multiplexed protein detection. By conjugating At1g60730 antibodies to isotopically pure metals instead of fluorophores, researchers can simultaneously detect dozens of proteins without spectral overlap concerns. This technology is increasingly being adapted for plant systems, offering unprecedented multiplexing capabilities for complex protein network analysis.

Basic Questions on Research Trends

Q: How are new antibody formats improving At1g60730 detection specificity and sensitivity?

Recent advances in antibody engineering have generated novel formats with enhanced properties for plant protein detection. Single-chain variable fragments (scFvs), which contain only the antigen-binding domains of conventional antibodies, offer improved tissue penetration for immunohistochemistry applications while maintaining target specificity. These smaller formats are particularly valuable for detecting proteins in dense plant tissues where antibody accessibility is limited.

Recombinant antibody technology is transforming plant protein detection by enabling precise antibody engineering and consistent production. Unlike traditional hybridoma-derived antibodies, recombinant antibodies are produced from cloned antibody genes, ensuring batch-to-batch consistency and eliminating the variability inherent in animal-derived antibodies. This approach addresses a fundamental challenge in antibody research, where "polyclonal antibodies remain widely used, necessitating alternative technologies" .

Nanobodies derived from camelid heavy-chain antibodies represent a revolutionary advance applicable to plant protein detection. These exceptionally small antibody fragments (approximately 15 kDa) exhibit remarkable stability and epitope recognition capabilities. Recent research demonstrates that "nanobodies are the best and most potently neutralizing antibodies to date" in certain applications, suggesting their potential value for detecting challenging plant targets like At1g60730.

Bispecific antibodies, which simultaneously bind two distinct epitopes, offer unprecedented specificity for complex plant samples. By requiring dual epitope recognition for binding, these engineered antibodies dramatically reduce cross-reactivity with related proteins. This approach particularly benefits Arabidopsis research, where protein families often contain multiple homologous members with high sequence similarity.

Q: What alternative technologies are emerging as complements or replacements for traditional antibodies in At1g60730 detection?

Emerging technologies are expanding the researcher's toolkit beyond traditional antibodies, offering complementary or alternative approaches for At1g60730 detection. Aptamers—short, single-stranded DNA or RNA molecules selected for target binding—represent a fully synthetic alternative to antibodies. These molecules can be generated through systematic evolution of ligands by exponential enrichment (SELEX), without requiring animal immunization, and offer exceptional stability under harsh conditions often encountered in plant research.

CRISPR-based protein detection systems like SHERLOCK (Specific High-sensitivity Enzymatic Reporter unLOCKing) adapt CRISPR-Cas technology for specific protein recognition. While still emerging for plant applications, these systems offer programmable recognition capabilities that could eventually complement antibody-based detection for proteins like At1g60730.

Proximity-based labeling technologies such as BioID or APEX2 enable detection of protein interactions and localization without requiring antibodies during the experimental phase. By fusing these enzymes to At1g60730, researchers can identify proximal proteins through biotinylation, with detection performed using standard streptavidin reagents rather than protein-specific antibodies.

Affimers and other non-antibody scaffolds represent an expanding class of engineered binding proteins with antibody-like specificity but improved stability and production consistency. These technologies address fundamental limitations of traditional antibodies, particularly for challenging research contexts where "despite these drawbacks, polyclonal antibodies remain widely used" due to lack of alternatives.

Q: How can computational approaches improve antibody selection and application for At1g60730 research?

Computational tools are revolutionizing antibody selection and application strategies for plant protein research. Epitope prediction algorithms analyze At1g60730 protein sequence and structure to identify optimal antigenic regions for antibody generation. These tools incorporate parameters including surface accessibility, hydrophilicity, and sequence conservation across related proteins to predict epitopes most likely to yield specific antibodies. By focusing antibody development on computationally validated epitopes, researchers dramatically improve success rates.

Structural modeling approaches, including protein threading and homology modeling, generate three-dimensional models of At1g60730 protein structure when crystallographic data is unavailable. These models enable visualization of potential epitope locations and accessibility, informing both antibody selection and experimental design. Molecular docking simulations further predict antibody-antigen binding interfaces, providing insights into binding affinity and specificity.

Machine learning algorithms trained on antibody performance data increasingly predict cross-reactivity profiles and optimal applications for existing antibodies. By analyzing sequence homology between At1g60730 and other plant proteins, these systems identify potential cross-reactive targets and suggest experimental conditions that maximize specificity. This approach is particularly valuable in plant systems with large protein families containing similar members.

Network analysis tools integrate antibody-based experimental data with transcriptomic and proteomic datasets, placing At1g60730 within broader biological contexts. These computational frameworks help researchers interpret antibody-derived localization or interaction data within comprehensive cellular pathways, enhancing biological relevance of individual findings.

Advanced Research Frontiers

Q: How can nanobody technology be applied to At1g60730 detection in challenging experimental contexts?

Nanobody technology offers transformative potential for At1g60730 detection in challenging experimental scenarios that limit conventional antibody effectiveness. These small, single-domain antibody fragments derived from camelid heavy-chain antibodies exhibit exceptional stability and epitope recognition properties that address fundamental limitations of traditional antibodies in plant research.

The remarkably small size of nanobodies (approximately 15 kDa, compared to 150 kDa for conventional antibodies) enables superior penetration into dense plant tissues and access to epitopes in crowded cellular environments. This characteristic is particularly valuable for detecting membrane-associated or cell wall-proximal proteins that might be inaccessible to larger antibodies. Research demonstrates that nanobodies can "mimic the recognition of the CD4 receptor—a key player in HIV infection" , suggesting their ability to access challenging epitopes.

Enhanced stability under demanding conditions represents another significant advantage of nanobodies for plant research. Unlike conventional antibodies, nanobodies retain functionality at elevated temperatures, in the presence of detergents, and across a wide pH range—conditions often encountered during plant sample processing. This stability enables detection protocols impossible with conventional antibodies, such as in situ analysis under harsh extraction conditions.

The modular nature of nanobodies facilitates creation of multivalent detection reagents through "engineering the nanobodies into a triple tandem format—by repeating short lengths of DNA" . For At1g60730 detection, researchers could generate nanobody constructs targeting multiple epitopes simultaneously, dramatically enhancing specificity and avidity. Moreover, nanobodies can be readily fused to fluorescent proteins or enzymes, creating direct detection reagents that eliminate secondary antibody requirements.

Q: What emerging super-resolution microscopy techniques are most compatible with At1g60730 antibody-based detection?

Super-resolution microscopy techniques are breaking the diffraction barrier that historically limited optical resolution to approximately 200 nm, enabling unprecedented visualization of At1g60730 localization and interactions at the nanoscale. Different super-resolution modalities offer complementary advantages for antibody-based detection in plant systems.

Stimulated Emission Depletion (STED) microscopy achieves resolution down to 30-80 nm by using a depletion laser to selectively suppress fluorescence emission around the periphery of the excitation spot. This technique is particularly compatible with standard immunofluorescence protocols using At1g60730 antibodies, requiring only specialized fluorophores with appropriate photophysical properties. STED offers the advantage of direct imaging without extensive post-processing, making it accessible for many plant biology laboratories.

Single-molecule localization microscopy techniques, including Photoactivated Localization Microscopy (PALM) and Stochastic Optical Reconstruction Microscopy (STORM), achieve 10-20 nm resolution by precisely localizing individual fluorophores across thousands of sequential images. These approaches require specialized photoactivatable or photoswitchable fluorophores conjugated to secondary antibodies, but offer exceptional resolution for revealing nanoscale distribution patterns of At1g60730 within subcellular compartments.

Expansion Microscopy (ExM) physically enlarges specimens through embedding in a swellable polymer network, achieving effective super-resolution with standard confocal microscopy. This approach is particularly valuable for plant tissues, as it simultaneously improves antibody accessibility by creating space between densely packed cellular components. When combined with At1g60730 antibodies, ExM can reveal previously unobservable spatial relationships between the target protein and cellular structures.

Q: How can antibody-based proteomics approaches be leveraged to study At1g60730 in systems biology contexts?

Antibody-based proteomics approaches enable systematic investigation of At1g60730 within comprehensive biological networks, bridging molecular-scale observations to system-level understanding. Reverse-phase protein arrays (RPPAs) represent a high-throughput platform for quantifying At1g60730 abundance across hundreds of samples simultaneously. By immobilizing plant extracts on nitrocellulose slides and probing with validated At1g60730 antibodies, researchers can analyze protein expression changes across developmental stages, tissues, or experimental conditions with exceptional statistical power.

Antibody-based proximity labeling approaches like proximity-dependent biotin identification (BioID) or enzyme-mediated activation of radical sources (APEX) enable comprehensive mapping of At1g60730's protein interaction network. By fusing promiscuous labeling enzymes to At1g60730, researchers can biotinylate proteins in close proximity, followed by streptavidin pulldown and mass spectrometry identification. This unbiased approach reveals both stable and transient interactions within their native cellular context.

Tissue microarrays combined with automated immunohistochemistry enable systematic analysis of At1g60730 expression patterns across multiple tissues and developmental stages. This approach parallels medical research methodologies, where antibodies like anti-Aurora-A kinase are used for "evaluation of Aurora A kinase expression in HeLa cells during mitosis" , adapting similar high-throughput analysis to plant systems.

Integration of antibody-derived data with transcriptomic and metabolomic datasets through computational modeling creates multi-dimensional understanding of At1g60730 function. Network analysis algorithms identify regulatory relationships and functional modules containing At1g60730, while machine learning approaches predict phenotypic outcomes based on protein expression patterns. These integrative approaches transform isolated antibody-based observations into comprehensive systems understanding.

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.