DHDPS2 Antibody

Shipped with Ice Packs
In Stock

Description

Key Features:

  • Gene Structure:

    • Plant DHDPS enzymes are encoded by multigene families. For example, Gm.DHDPS-B in soybeans shows distinct expression patterns compared to Gm.DHDPS-A1/A2 .

    • Evolutionary analysis reveals conserved catalytic domains across species, with plant-specific structural adaptations .

Research Findings on DHDPS-B (Putative DHDPS2)

Recent studies highlight the role of DHDPS-B in stress responses and metabolic regulation:

A. Expression Under Abiotic and Biotic Stress

  • Ethylene Treatment:

    ConditionGm.DHDPS-B Log2Fold ChangeSignificance (p-value)
    Leaf petiole (24 h)+4.0<0.05
    Leaf petiole (48 h)+4.1<0.05
    • Upregulation suggests a role in ethylene-mediated stress signaling .

  • Salt Stress:

    • Significant downregulation (−3.5 Log2Fold change) in roots, though not statistically significant .

B. Comparative Expression Across Tissues

  • Baseline Activity:

    TissueGm.DHDPS-B RPKMGm.DHDPS-A1/A2 RPKM
    Germination-stage root1.940.21–0.56
    Senescent leaf0.530.12–0.45
    • Gm.DHDPS-B exhibits tissue-specific expression, contrasting with ubiquitous DHDPS-A isoforms .

Antibody Development and Detection

While no studies directly describe "DHDPS2 Antibody," methodologies for targeting DHDPS isoforms can be inferred:

A. Antibody Generation Strategies

  • Epitope Selection:

    • Antibodies against conserved catalytic domains (e.g., residues 80–120 in Gm.DHDPS-B) enable isoform-specific detection .

  • Validation:

    • Western blotting and ELISA are standard for confirming antibody specificity, as seen in analogous studies on anti-DSG2 antibodies .

B. Applications in Research

  • Stress Response Studies:

    • Antibodies facilitate tracking DHDPS-B dynamics under stress (e.g., flooding, ozone exposure) .

  • Localization:

    • Immunofluorescence reveals subcellular distribution, such as plastid localization in plant cells .

Comparative Analysis with Other Antibodies

FeatureAnti-DHDPS-B (Hypothetical)Anti-DSG2 (ARVC Study) Anti-OSP (Cholera Study)
TargetMetabolic enzymeCardiac desmosome proteinBacterial polysaccharide
Clinical RelevanceCrop stress resistanceArrhythmia diagnosisPathogen neutralization
Detection MethodWestern blot, ELISAIFL, ELISAAgglutination assays

Unresolved Questions and Future Directions

  • Functional Redundancy: The interplay between DHDPS-A and DHDPS-B isoforms under stress remains unclear .

  • Therapeutic Potential: No studies explore DHDPS antibodies in human health, though plant-focused applications (e.g., crop yield optimization) are emerging .

Product Specs

Buffer
Preservative: 0.03% ProClin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
14-16 weeks (made-to-order)
Synonyms
DHDPS2 antibody; At2g45440 antibody; F4L23.54-hydroxy-tetrahydrodipicolinate synthase 2 antibody; chloroplastic antibody; HTPA synthase 2 antibody; EC 4.3.3.7 antibody
Target Names
DHDPS2
Uniprot No.

Target Background

Function
This antibody targets DHDPS2, an enzyme that catalyzes the condensation of (S)-aspartate-β-semialdehyde [(S)-ASA] and pyruvate to form 4-hydroxy-tetrahydrodipicolinate (HTPA).
Gene References Into Functions

Further insights into DHDPS2 function are provided by these studies:

  1. Structural studies of Arabidopsis thaliana DHDPS2 (At-DHDPS2) reveal an allosteric site for (S)-lysine binding within a cleft between two monomers. PMID: 22792278
  2. This publication details the cloning, expression, purification, crystallization, and preliminary X-ray diffraction analysis of At-DHDPS2. Diffraction-quality crystals were obtained, belonging to space group P2(1)2(1)2. PMID: 22102238
Database Links

KEGG: ath:AT2G45440

STRING: 3702.AT2G45440.1

UniGene: At.12311

Protein Families
DapA family
Subcellular Location
Plastid, chloroplast.

Q&A

What is DHDPS2 and why is it important for plant research?

DHDPS2 (Dihydrodipicolinate synthase 2) is a key enzyme in the aspartate-derived lysine biosynthesis pathway in plants. It catalyzes the condensation of pyruvate and aspartate-β-semialdehyde to form 2,3-dihydrodipicolinate, which is the first committed step in lysine biosynthesis. This enzyme is critical for several reasons:

  • It represents a rate-limiting step in lysine production, making it an important target for biofortification strategies aimed at increasing essential amino acid content in crops .

  • DHDPS enzymes show tissue-specific expression patterns, with different isoforms (such as DHDPS-A and DHDPS-B types) being expressed in different plant tissues and developmental stages .

  • The enzyme is responsive to various environmental stresses, potentially playing roles in plant adaptation mechanisms .

Research on DHDPS2 contributes to our understanding of plant metabolism, protein synthesis, and stress responses, with potential applications in crop improvement and nutritional enhancement.

How can I select the appropriate DHDPS2 antibody for my research?

Selecting the appropriate DHDPS2 antibody requires careful consideration of several factors:

  • Specificity: Determine whether the antibody is specific to DHDPS2 or cross-reacts with other DHDPS isoforms. Review validation data showing discrimination between DHDPS1, DHDPS2, and other related proteins.

  • Application compatibility: Verify that the antibody has been validated for your specific application (Western blotting, immunohistochemistry, immunoprecipitation, ELISA, etc.) .

  • Species reactivity: Confirm that the antibody recognizes DHDPS2 from your species of interest. Plant DHDPS2 sequences can vary between species, affecting epitope recognition .

  • Epitope information: Antibodies raised against different regions of DHDPS2 may perform differently. Those targeting unique regions are more likely to be isoform-specific.

  • Validation methods: Review how the antibody was validated. Ideally, validation should include positive controls (recombinant DHDPS2 protein), negative controls (knockout or knockdown samples), and specificity tests against related proteins.

A methodical approach to antibody selection increases the likelihood of obtaining reliable and reproducible results in your DHDPS2 research.

What are the best methods for validating a DHDPS2 antibody?

Validating a DHDPS2 antibody thoroughly is essential for ensuring reliable experimental results. Recommended validation methods include:

  • Western blot with recombinant protein: Test the antibody against purified recombinant DHDPS2 protein to confirm binding and determine sensitivity.

  • Western blot with positive and negative tissue samples: Use tissues known to express high levels of DHDPS2 (based on transcriptomic data) as positive controls and tissues with low or no expression as negative controls .

  • Knockout/knockdown validation: If available, test the antibody on samples from DHDPS2 knockout or knockdown plants. The specific band should be absent or significantly reduced.

  • Peptide competition assay: Pre-incubate the antibody with the immunizing peptide before application. This should block specific binding and eliminate the DHDPS2 signal.

  • Cross-reactivity assessment: Test against recombinant DHDPS1 and other related proteins to ensure specificity for DHDPS2.

  • Immunohistochemistry correlation: Compare immunohistochemistry results with in situ hybridization or promoter-reporter studies to verify whether protein localization matches expression patterns.

Thorough validation using multiple approaches provides confidence in antibody specificity and performance across different experimental conditions.

What are the typical expression patterns of DHDPS2 in plant tissues?

DHDPS2 expression varies across plant tissues and developmental stages, making it important to understand these patterns when designing experiments. Based on RNA-seq data analysis of DHDPS genes in soybean (Glycine max):

  • Seed expression: DHDPS-A type genes (analogous to DHDPS2 in some species) show highest expression in developing seeds, particularly at 42 days after flowering (DAF) with RPKM values of 4.26 and 4.36 for the two A-type isoforms .

  • Root expression: DHDPS-B type shows highest expression in roots at the germination stage (RPKM of 1.94) compared to other tissues .

  • Developmental regulation: Expression patterns change throughout development, with the second-highest DHDPS-B expression found in cotyledons at the trefoil stage (RPKM of 1.31) but not detected in cotyledons at the germination stage .

  • Stem and leaf expression: Low but detectable DHDPS-B expression occurs in stems at germination stage and in senescent leaves (RPKM values of 0.56 and 0.53, respectively) .

This tissue-specific expression pattern suggests that different DHDPS isoforms may have specialized functions in different plant tissues and developmental stages.

TissueDHDPS-A1 RPKMDHDPS-A2 RPKMDHDPS-B RPKM
Seeds (42 DAF)4.264.36Low/not detected
Roots (germination)LowLow1.94
Cotyledon (trefoil)LowLow1.31
Stem (germination)LowLow0.56
Senescent leavesLowLow0.53

How does DHDPS2 expression change under various stress conditions?

DHDPS2 expression exhibits differential responses to various abiotic and biotic stresses, providing insights into its potential roles in stress adaptation. Analysis of multiple RNA-seq datasets reveals:

  • Ethylene treatment: Significant differential expression of DHDPS genes occurs following ethylene treatment. While DHDPS-A type genes (analogous to DHDPS2 in some species) are generally downregulated, DHDPS-B type shows strong upregulation in leaf petioles after 24h and 48h of ethylene treatment (Log2Fold changes of 4.0 and 4.1, respectively, p < 0.05) .

  • Salt stress: DHDPS-B type shows substantial downregulation in leaves after 1h of salt treatment (Log2Fold change of -3.5), although this change was not statistically significant (p > 0.05) .

  • Viral infection: After SMV (Soybean Mosaic Virus) treatment, DHDPS-B exhibits strong downregulation at 12h and 24h post-infection (Log2Fold changes of -5.0 and -4.8, respectively), though these changes were not statistically significant (p > 0.05) .

  • Other abiotic stresses: DHDPS genes show significant responses to flooding, water deficit, ozone, and some salt treatments, with DHDPS-B generally being upregulated in these conditions .

These expression changes suggest that DHDPS enzymes may play roles beyond basic lysine biosynthesis, potentially contributing to stress adaptation mechanisms.

Stress ConditionTime PointDHDPS-A1 Log2FCDHDPS-A2 Log2FCDHDPS-B Log2FCStatistical Significance
Ethylene (leaf petiole)24hDownregulatedDownregulated+4.0p < 0.05
Ethylene (leaf petiole)48hDownregulatedDownregulated+4.1p < 0.05
Salt (leaves)1hMinimal changeMinimal change-3.5Not significant (p > 0.05)
SMV infection12hMinimal changeMinimal change-5.0Not significant (p > 0.05)
SMV infection24hMinimal changeMinimal change-4.8Not significant (p > 0.05)

What are the challenges in developing highly specific antibodies against DHDPS2?

Developing highly specific antibodies against DHDPS2 presents several challenges that researchers should consider:

  • Homology with related isoforms: DHDPS1 and DHDPS2 typically share significant sequence homology, making it difficult to identify unique epitopes that distinguish between isoforms. This challenge is similar to those faced when developing antibodies against other closely related protein families .

  • Conservation across species: If developing antibodies for use across multiple plant species, the high conservation of functional domains in DHDPS enzymes can limit the regions suitable for species-specific antibody development.

  • Post-translational modifications: DHDPS2 may undergo post-translational modifications that affect epitope accessibility or alter the protein conformation, affecting antibody recognition.

  • Native protein structure: The native folding of DHDPS2 may mask linear epitopes that are accessible in denatured conditions, creating differences in antibody performance between applications (e.g., Western blot versus immunoprecipitation).

  • Expression levels: Relatively low expression levels of DHDPS2 in some tissues may require antibodies with particularly high affinity and sensitivity .

Overcoming these challenges requires careful epitope selection, comprehensive validation, and potentially the use of computational approaches to design antibodies with enhanced specificity .

How can computational models help design more specific DHDPS2 antibodies?

Computational approaches can significantly enhance the design of specific DHDPS2 antibodies, addressing many challenges in traditional antibody development:

  • Epitope prediction and selection: Advanced algorithms can identify unique epitopes in DHDPS2 that have minimal homology with DHDPS1 and other related proteins, increasing the likelihood of isoform-specific antibody development.

  • Structural modeling: Protein structure prediction tools can model the three-dimensional structure of DHDPS2, identifying surface-exposed regions that are accessible for antibody binding in the native protein.

  • Machine learning approaches: Variational autoencoder (VAE) models and other deep learning approaches can analyze antibody-antigen interactions to predict binding properties . These models can:

    • Generate novel antibody sequences with desired specificity profiles

    • Identify antibody sequences likely to bind specific epitopes with high affinity

    • Predict cross-reactivity with related proteins

  • Convergent selection analysis: By analyzing natural antibody repertoires, computational methods can identify convergent patterns in antibody sequences that recognize specific antigens, informing rational antibody design .

  • Biophysics-informed modeling: Combining experimental data with biophysical principles can identify different binding modes associated with particular ligands, enabling the computational design of antibodies with customized specificity profiles .

For example, researchers have demonstrated that VAE models trained on antibody repertoire sequencing data can successfully predict binding properties of novel antibody variants, even those not present in the training data .

What methods can be used to analyze post-translational modifications of DHDPS2?

Analyzing post-translational modifications (PTMs) of DHDPS2 requires specialized techniques that often incorporate antibody-based approaches:

  • Immunoprecipitation coupled with mass spectrometry:

    • Use a validated DHDPS2 antibody to immunoprecipitate the protein from plant extracts

    • Analyze the purified protein by liquid chromatography-tandem mass spectrometry (LC-MS/MS)

    • This method can identify various PTMs including phosphorylation, acetylation, ubiquitination, and glycosylation

  • Phospho-specific antibodies:

    • Develop antibodies specifically targeting phosphorylated residues of DHDPS2

    • These can be used to monitor phosphorylation status under different conditions

    • Requires prediction or prior knowledge of phosphorylation sites

  • 2D gel electrophoresis with Western blotting:

    • Separate proteins based on both isoelectric point and molecular weight

    • Detect DHDPS2 using specific antibodies

    • PTMs often cause shifts in isoelectric point or apparent molecular weight

  • Phos-tag SDS-PAGE:

    • A modified SDS-PAGE technique that can separate phosphorylated from non-phosphorylated proteins

    • When combined with Western blotting using DHDPS2 antibodies, it can detect phosphorylated forms

  • Site-directed mutagenesis coupled with functional assays:

    • Create mutations at potential PTM sites

    • Compare activity and localization with wild-type protein

    • Use antibodies to assess how mutations affect protein stability or interactions

These methods, particularly when used in combination, can provide comprehensive insights into how PTMs regulate DHDPS2 function, localization, and interactions in response to developmental or environmental cues.

How can CRISPR-Cas9 be used to study DHDPS2 function and validate antibodies?

CRISPR-Cas9 technology offers powerful approaches for studying DHDPS2 function and generating valuable resources for antibody validation:

  • Generation of knockout lines:

    • Design guide RNAs targeting DHDPS2-specific exons

    • Create complete knockout plants lacking DHDPS2 expression

    • These plants serve as perfect negative controls for antibody validation, as any signal detected in knockout tissues would indicate non-specific binding

    • Phenotypic analysis of knockouts can reveal DHDPS2's physiological roles

  • CRISPR-Cas9 homology-directed mutagenesis:

    • Introduce precise mutations in the DHDPS2 gene using homology-directed repair templates

    • Create variants with altered catalytic activity or regulation

    • Study how specific amino acid changes affect enzyme function and plant phenotype

  • Epitope tagging:

    • Use CRISPR-Cas9 to add epitope tags (HA, FLAG, etc.) to the endogenous DHDPS2 gene

    • This allows detection of the protein at endogenous levels using well-validated commercial tag antibodies

    • Provides a way to study DHDPS2 when specific antibodies are unavailable or problematic

  • Creation of antibody validation libraries:

    • Generate libraries of DHDPS2 variants through CRISPR-Cas9 homology-directed mutagenesis

    • Screen variants for antibody binding

    • Identify critical residues for antibody recognition

    • This approach can help characterize antibody specificity and potential cross-reactivity

CRISPR-Cas9 approaches not only provide essential validation tools for DHDPS2 antibodies but also create experimental systems to understand the protein's function, regulation, and importance in plant physiology.

What are the optimal protein extraction methods for DHDPS2 detection by Western blot?

Effective protein extraction is crucial for reliable DHDPS2 detection in Western blot analysis. Consider these optimization strategies:

  • Buffer composition:

    • Start with a standard plant protein extraction buffer containing:

      • 50 mM Tris-HCl (pH 7.5)

      • 150 mM NaCl

      • 1% Triton X-100 or NP-40

      • 1 mM EDTA

    • Add protease inhibitor cocktail to prevent degradation

    • Include phosphatase inhibitors if studying phosphorylation states

  • Tissue-specific considerations:

    • For seeds and other storage tissues, add 1-2% SDS to improve protein extraction

    • For leaves and green tissues, add 1-2% PVPP to remove interfering phenolic compounds

    • For tissues with high starch content, consider additional centrifugation steps

  • Protein fractionation approaches:

    • Sequential extraction can help identify subcellular localization:

      • Extract soluble proteins first with non-ionic detergent buffer

      • Then extract membrane-associated proteins with stronger detergents

      • Finally, extract nuclear proteins with high-salt buffers

    • Compare DHDPS2 distribution across fractions using antibody detection

  • Denaturation conditions:

    • Test multiple sample heating conditions (65°C, 95°C, and non-boiled)

    • Some proteins aggregate when boiled, affecting detection

    • Include reducing agents (DTT or β-mercaptoethanol) to break disulfide bonds

  • Loading controls:

    • Use antibodies against housekeeping proteins appropriate for your tissue type

    • Consider using total protein staining methods (Ponceau S, Coomassie, SYPRO Ruby) as loading controls

A systematically optimized protein extraction protocol ensures reliable and reproducible detection of DHDPS2 by Western blot, enabling accurate quantification of protein levels across different experimental conditions.

How can I optimize immunohistochemistry protocols for DHDPS2 localization studies?

Optimizing immunohistochemistry (IHC) protocols for DHDPS2 localization requires attention to several critical parameters:

  • Tissue fixation:

    • Test multiple fixatives: 4% paraformaldehyde, Carnoy's solution, and glutaraldehyde

    • Optimize fixation time (typically 4-24 hours) to balance tissue preservation and epitope accessibility

    • Consider perfusion fixation for whole plant seedlings when possible

  • Antigen retrieval methods:

    • Heat-induced epitope retrieval (HIER): Test different buffers (citrate pH 6.0, EDTA pH 8.0) and heating times

    • Enzymatic retrieval: Try proteinase K or trypsin digestion at varying concentrations and incubation times

    • Some epitopes may require specific retrieval methods for optimal detection

  • Blocking and permeabilization:

    • Test different blocking agents: BSA (3-5%), normal serum (5-10%), or commercial blocking reagents

    • Optimize permeabilization with detergents (0.1-0.5% Triton X-100 or 0.05-0.1% Tween-20)

    • Consider a separate permeabilization step before blocking for thick sections

  • Antibody incubation conditions:

    • Test different antibody dilutions (typically 1:50 to 1:500 for IHC)

    • Compare overnight incubation at 4°C versus shorter incubations at room temperature

    • Evaluate whether adding detergents (0.01-0.05% Tween-20) to antibody solutions improves results

  • Controls and validation:

    • Include tissue from DHDPS2 knockout or knockdown plants as negative controls

    • Use preimmune serum controls and secondary-only controls to assess background

    • Perform peptide competition assays to confirm specificity

    • Consider dual labeling with markers of known subcellular compartments

A systematic approach to optimizing these parameters will provide reliable and reproducible DHDPS2 localization data, enabling insights into its subcellular distribution and potential functional associations.

What approaches can be used to quantify DHDPS2 protein levels accurately?

Accurate quantification of DHDPS2 protein levels is essential for comparative studies across different conditions or genotypes. Several complementary approaches can be employed:

  • Quantitative Western blotting:

    • Use internal loading controls (housekeeping proteins or total protein staining)

    • Include a standard curve of recombinant DHDPS2 protein

    • Employ digital image analysis software for densitometry

    • Consider fluorescent secondary antibodies for wider linear range than chemiluminescence

    • Present data as relative values normalized to controls

  • ELISA (Enzyme-Linked Immunosorbent Assay):

    • Develop sandwich ELISA using two different DHDPS2 antibodies recognizing distinct epitopes

    • Create a standard curve using purified recombinant DHDPS2

    • This method allows high-throughput analysis of multiple samples

    • Typically provides better quantitative accuracy than Western blotting

  • Mass spectrometry-based approaches:

    • Selected Reaction Monitoring (SRM) or Multiple Reaction Monitoring (MRM)

    • Parallel Reaction Monitoring (PRM)

    • These targeted approaches can quantify DHDPS2 with high specificity

    • Use isotope-labeled peptides as internal standards

    • Can simultaneously quantify multiple proteins, including different DHDPS isoforms

  • Data analysis considerations:

    • Use biological and technical replicates (minimum n=3)

    • Apply appropriate statistical tests

    • Consider normalization to total protein rather than single reference proteins

    • Report both absolute and relative quantification when possible

MethodAdvantagesLimitationsBest For
Western blottingWidely accessible, visualizes protein sizeSemi-quantitative, variable linearityComparing relative levels, detecting isoforms
ELISAHigh throughput, good quantitative accuracyRequires two antibodies, no size informationPrecise quantification across many samples
Targeted MSHigh specificity, absolute quantificationRequires specialized equipment, complex setupMultiplex analysis, absolute quantification
IP-based methodsEnhanced sensitivityLabor-intensive, potential bias in precipitationLow-abundance proteins

How should I interpret contradictory results when using different DHDPS2 antibodies?

Resolving contradictory results obtained with different DHDPS2 antibodies requires systematic troubleshooting and validation:

  • Epitope mapping:

    • Determine the specific regions of DHDPS2 recognized by each antibody

    • Different epitopes may be differentially accessible in various experimental conditions

    • Some epitopes may be masked by protein-protein interactions or post-translational modifications

  • Validation with multiple techniques:

    • Compare results across different applications (Western blot, immunohistochemistry, ELISA)

    • Inconsistencies across techniques may reveal condition-specific epitope accessibility

  • Use of genetic controls:

    • Test antibodies on samples from DHDPS2 knockout or knockdown plants

    • Any signal in these negative controls indicates non-specific binding

    • Overexpression lines can serve as positive controls

  • Cross-reactivity assessment:

    • Test antibodies against recombinant DHDPS1 and other related proteins

    • Determine if contradictory results stem from differential cross-reactivity

  • Antibody validation with modified DHDPS2 variants:

    • Generate DHDPS2 variants with mutations in epitope regions

    • Test antibody binding to identify critical recognition residues

    • This can help interpret contradictory results if some variants bind one antibody but not others

When reporting results, transparently document which antibody was used for each experiment and acknowledge any discrepancies between antibodies, providing possible explanations based on your validation studies.

What controls are essential for publication-quality DHDPS2 antibody research?

For publication-quality research using DHDPS2 antibodies, the following controls are essential:

  • Specificity controls:

    • Genetic knockouts or knockdowns of DHDPS2

    • Peptide competition assays (pre-incubation of antibody with immunizing peptide)

    • Preimmune serum controls (for polyclonal antibodies)

    • Isotype controls (for monoclonal antibodies)

    • Recombinant protein controls (positive control)

  • Technical controls:

    • Secondary antibody-only controls to assess non-specific binding

    • Loading controls for Western blots (housekeeping proteins or total protein staining)

    • Tissue processing controls (processing samples identically)

    • Multiple antibody dilutions to demonstrate signal specificity

    • Inclusion of related proteins (e.g., DHDPS1) to demonstrate isoform specificity

  • Biological controls:

    • Tissues known to express high levels of DHDPS2 (positive biological control)

    • Tissues with low/no expression of DHDPS2 (negative biological control)

    • Multiple biological replicates (minimum n=3)

    • Samples representing different developmental stages or conditions

  • Validation across methods:

    • Confirmation of key findings using at least two independent methods

    • Correlation between protein detection and mRNA expression data

    • Complementary approaches (e.g., epitope-tagged constructs)

  • Quantification controls:

    • Standard curves for quantitative analyses

    • Technical replicates for quantification

    • Appropriate statistical analysis

    • Concentration-dependent response demonstration

Rigorous use of these controls ensures that research findings are robust, reproducible, and correctly interpreted, meeting the high standards required for publication in peer-reviewed journals.

What are the future research directions for DHDPS2 antibody development and applications?

Future research on DHDPS2 antibodies and their applications will likely focus on several promising directions:

  • Advanced computational design approaches:

    • Integration of deep learning methods to design highly specific antibodies targeting unique DHDPS2 epitopes

    • Development of in silico prediction tools to optimize antibody-antigen binding and minimize cross-reactivity

    • Application of structure-based design to engineer antibodies with enhanced binding properties

  • Isoform-specific antibody development:

    • Creation of antibodies that can reliably distinguish between DHDPS1 and DHDPS2, as well as between A-type and B-type isoforms

    • Development of antibodies specific to particular post-translationally modified forms of DHDPS2

    • Production of antibodies targeting species-specific variants of DHDPS2

  • Single-cell and in vivo applications:

    • Development of antibody-based biosensors for real-time monitoring of DHDPS2 in living plant cells

    • Application of DHDPS2 antibodies in single-cell proteomics approaches

    • Creation of intrabodies that can be expressed within plant cells to track or modulate DHDPS2 function

  • High-throughput screening platforms:

    • Development of antibody-based assays for screening plant varieties with altered DHDPS2 levels or activity

    • Creation of biosensor platforms to detect DHDPS2 responses to environmental stresses

    • Establishment of automated systems for rapid DHDPS2 quantification across large sample sets

  • Integration with CRISPR technologies:

    • Continued refinement of CRISPR-based approaches for antibody validation

    • Development of CRISPR knock-in systems for epitope tagging endogenous DHDPS2

    • Creation of antibody validation libraries through CRISPR-Cas9 homology-directed mutagenesis

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.