The AT5G58784 gene product belongs to the UPPS family, which catalyzes the synthesis of undecaprenyl pyrophosphate—a lipid carrier critical for cell wall biosynthesis in bacteria and plants. In Arabidopsis, this protein is hypothesized to play a role in glycosylation pathways or cell wall remodeling, though functional studies are sparse .
| Gene Identifier | Annotation | Species | Protein Class |
|---|---|---|---|
| AT5G58784 | UPPS family protein | Arabidopsis thaliana | Undecaprenyl pyrophosphate synthetase |
While no commercial or peer-reviewed studies explicitly describe an "AT5G58784 antibody," antibodies targeting plant proteins typically follow these workflows:
Immunogen Design: Peptides or recombinant proteins derived from the target sequence are used for immunization .
Validation: Western blot, ELISA, or immunohistochemistry confirm specificity, often using knockout mutants as negative controls .
Applications: Localization studies, protein interaction assays, or functional knockdown experiments .
For AT5G58784, hypothetical applications might include:
Investigating its role in cell wall biosynthesis.
Studying subcellular localization in plant tissues.
The lack of AT5G58784-specific antibody data underscores broader issues in antibody reliability. Key considerations:
Specificity: Cross-reactivity with homologous UPPS proteins (e.g., bacterial homologs) must be ruled out .
Epitope Accessibility: Membrane-associated proteins like UPPS may require detergent-based extraction for antibody binding .
Current literature lacks direct evidence of AT5G58784 antibody use. Prioritizing the following could advance this field:
Recombinant Antibody Production: Phage display or hybridoma technology to generate high-affinity binders .
Functional Studies: Linking AT5G58784 to specific biochemical pathways using CRISPR-edited mutants.
Cross-Species Comparisons: Analyzing UPPS homologs in pathogenic bacteria for antibiotic development .
At5g58784 encodes a 35.1 dehydrodolichyl diphosphate synthase in Arabidopsis thaliana, which plays a crucial role in isoprenoid biosynthesis and post-translational protein modifications . This enzyme catalyzes the formation of dehydrodolichyl diphosphate, a precursor in dolichol biosynthesis that is essential for glycoprotein processing. The protein's function is particularly relevant in studies of plant stress responses, as transcriptomic data indicates differential expression under salinity conditions .
Methodological approach: When studying this protein, researchers should consider both transcriptional analysis (qRT-PCR, RNA-seq) and protein-level investigations using antibody-based techniques. Comparative analysis with homologous proteins in other plant species can provide evolutionary insights into function conservation.
For optimal detection of the At5g58784 protein using antibodies, follow these methodological steps:
Tissue homogenization: Grind plant tissue in liquid nitrogen and extract with buffer containing 50 mM Tris-HCl (pH 7.5), 150 mM NaCl, 1% Triton X-100, and protease inhibitor cocktail
Clarification: Centrifuge at 12,000 × g for 30 minutes at 4°C, followed by filtration through 0.45μm and 0.22μm filters as performed in standard antibody preparation protocols
Protein quantification: Use Bradford or BCA assay to normalize protein concentrations
Denaturation: For western blotting, denature samples in Laemmli buffer at 95°C for 5 minutes
Fractionation: Consider subcellular fractionation to enrich for membrane fractions where this enzyme is likely to localize
The quality of sample preparation directly impacts antibody detection specificity. For challenging plant tissues with high polysaccharide or secondary metabolite content, additional purification steps may be necessary.
Validation of At5g58784 antibody specificity requires a multi-technique approach similar to established antibody validation principles:
Western blot analysis using:
Wild-type Arabidopsis tissues
At5g58784 knockout/knockdown mutants (negative control)
Recombinant At5g58784 protein (positive control)
Analysis of band size (predicted molecular weight: approximately 35 kDa)
Immunoprecipitation followed by mass spectrometry identification
Immunofluorescence with appropriate controls, comparing localization patterns with predictions based on bioinformatic analysis
Always include technical replicates and appropriate controls, particularly knockout/knockdown lines when available. Cross-reactivity testing against related dehydrodolichyl diphosphate synthase family members is critical for confirming specificity.
Developing monoclonal antibodies against At5g58784 requires strategic epitope selection and meticulous validation:
Epitope prediction and selection:
Analyze hydrophilicity, surface accessibility, and sequence conservation
Avoid transmembrane domains and heavily glycosylated regions
Consider both linear and conformational epitopes
Antigen preparation options:
Recombinant full-length protein expression
Synthetic peptide conjugation to carrier proteins
Domain-specific constructs for increased solubility
Hybridoma development protocol:
Immunize mice with purified antigen following standard protocols
Perform cell fusion using polyethylene glycol method
Screen hybridoma supernatants using ELISA against the immunizing antigen
Confirm specificity using western blot, immunoprecipitation and immunofluorescence techniques
Consider using Fabrack-CAR technology for improved specificity in complex systems
Validation in multiple assays:
Western blotting
Immunoprecipitation
Immunofluorescence/immunohistochemistry
Flow cytometry (for single-cell plant suspensions)
For advanced applications, consider epitope tagging the endogenous At5g58784 gene using CRISPR/Cas9 genome editing to create a system for comparison and validation.
At5g58784 shows differential expression under salt stress conditions, as evidenced in plant transcriptional response studies . For quantitative assessment of these changes at the protein level:
Quantitative western blotting:
Use fluorescent secondary antibodies for wider dynamic range
Include reference proteins for normalization (constitutively expressed)
Employ time-course analysis (0, 6, 12, 24, 48, 72 hours post-stress)
Immunohistochemistry for tissue-specific localization:
Compare control vs. salt-stressed plants
Quantify signal intensity across different tissues
Co-localize with known stress response markers
Proteomics approach:
Immunoprecipitation combined with mass spectrometry
Analysis of post-translational modifications
Identification of interaction partners under stress conditions
ELISA-based quantification:
Develop sandwich ELISA using capture and detection antibodies
Generate standard curves with recombinant protein
Measure protein levels across different stress conditions
| Technique | Advantages | Limitations | Sample Requirements |
|---|---|---|---|
| Western blot | Direct visualization, size confirmation | Semi-quantitative | 20-50 μg total protein |
| ELISA | Higher throughput, quantitative | Requires two non-competing antibodies | 5-10 μg total protein |
| Immunohistochemistry | Spatial information | Qualitative, fixation artifacts | Fixed tissue sections |
| IP-MS | Interaction partners, PTMs | Complex analysis, expensive | 0.5-1 mg total protein |
For multiplex immunofluorescence experiments incorporating At5g58784 antibodies:
Antibody compatibility planning:
Select primary antibodies from different host species
If using same-species antibodies, consider direct labeling or sequential staining
Test for cross-reactivity between secondary antibodies
Validate each antibody individually before multiplexing
Signal separation strategies:
Technical optimization:
Determine optimal fixation method (paraformaldehyde vs. methanol)
Test different antigen retrieval methods
Titrate antibody concentrations to minimize background
Include appropriate blocking agents to reduce non-specific binding
Controls and validation:
Single-color controls for spectral compensation
Knockout/knockdown controls for specificity
Secondary-only controls for background assessment
Co-localization analysis with known organelle markers
When designing multiplex experiments, consider the biological relationship between targets and select markers that address your specific research question about At5g58784 function in relevant cellular compartments or processes.
When encountering signal problems with At5g58784 antibodies:
Sample preparation optimization:
Ensure complete protein extraction with appropriate buffer composition
Add phosphatase and protease inhibitors to preserve protein integrity
Test different reducing conditions (DTT vs. β-mercaptoethanol)
Optimize protein loading (typically 25-50 μg total protein)
Transfer efficiency improvement:
Adjust transfer conditions for high molecular weight proteins
Consider semi-dry vs. wet transfer methods
Validate transfer with reversible staining (Ponceau S)
Test PVDF vs. nitrocellulose membranes
Antibody incubation parameters:
Extend primary antibody incubation time (overnight at 4°C)
Test different antibody dilutions (typically 1:500 to 1:2000)
Add 0.1% Tween-20 or 0.05% Triton X-100 to reduce background
Consider more sensitive detection systems (ECL-Plus, fluorescent)
Epitope accessibility enhancement:
Test different antigen retrieval methods
Try native vs. denaturing conditions
Consider membrane treatment with methanol or SDS
For recalcitrant targets, consulting antibody data repositories may provide insights from other researchers' experiences with similar plant proteins .
To minimize cross-reactivity problems:
Specificity enhancement approaches:
Affinity purification of polyclonal antibodies against the specific antigen
Pre-adsorption with related proteins or tissue lysates from knockout plants
Use of monoclonal antibodies with defined epitope specificity
Consider competitive blocking with immunizing peptide
Assay-specific optimization:
Increase washing stringency (higher salt, detergent concentration)
Optimize blocking (5% BSA vs. 5% non-fat milk)
Adjust antibody concentration to minimize non-specific binding
Test different fixation protocols for immunohistochemistry
Validation in knockout systems:
Compare signal between wild-type and At5g58784 mutant plants
Use CRISPR/Cas9-generated knockout lines as definitive controls
Consider RNAi knockdown lines for partial expression reduction
Correlate protein levels with transcript abundance
Advanced specificity testing:
Peptide array epitope mapping
Competitive ELISA with related protein sequences
Mass spectrometry validation of immunoprecipitated proteins
Immunodepletion experiments
Careful antibody validation using multiple approaches is essential for distinguishing specific from non-specific signals, particularly in plant systems where antibody resources are more limited than in mammalian research.
For successful ChIP applications with At5g58784 antibodies:
Experimental design considerations:
Determine if At5g58784 has potential DNA-binding activity or associates with chromatin-modifying complexes
Design appropriate crosslinking conditions (1% formaldehyde, 10 minutes)
Optimize sonication parameters for 200-500 bp chromatin fragments
Include appropriate controls (IgG, input, non-target protein)
Protocol modifications for plant tissues:
Use vacuum infiltration for formaldehyde crosslinking
Consider nuclear isolation before sonication
Increase washing stringency to reduce plant-specific background
Test different antibody concentrations and incubation times
Validation approaches:
qPCR of known or predicted target regions
ChIP-seq for genome-wide binding profile
Compare results with transcriptomic data sets
Verify findings with alternative antibodies when available
Data analysis considerations:
Normalize to input controls
Use appropriate statistical methods for peak calling
Perform motif analysis on enriched regions
Integrate with other genomic datasets
If At5g58784 functions primarily as a metabolic enzyme rather than a DNA-binding protein, consider alternative approaches like ChIP-MS to identify potential chromatin-associated protein complexes containing At5g58784.
When employing At5g58784 antibodies in interaction studies:
Co-immunoprecipitation (Co-IP) optimization:
Test different lysis conditions to preserve interactions
Optimize antibody concentration and incubation time
Consider crosslinking for transient interactions
Use appropriate controls (IgG, knockout/knockdown lines)
Proximity ligation assay (PLA) implementation:
Select antibody pairs from different host species
Optimize fixation and permeabilization for plant tissues
Include appropriate controls for probe specificity
Quantify PLA signals across different experimental conditions
Immunofluorescence co-localization:
Use high-resolution microscopy (confocal, super-resolution)
Apply appropriate co-localization statistics (Pearson's, Mander's)
Consider live-cell imaging for dynamic interactions
Control for random co-localization events
Analytical considerations:
Confirm interactions with reciprocal IP when possible
Validate with orthogonal methods (Y2H, BiFC, FRET)
Consider changes in interaction dynamics under stress conditions
Analyze interaction domains through truncation constructs
| Method | Resolution | Quantification | In vivo/vitro | Technical Complexity |
|---|---|---|---|---|
| Co-IP | Protein complex | Semi-quantitative | In vitro | Medium |
| PLA | Single-molecule | Quantitative | In situ | High |
| Co-localization | Subcellular | Semi-quantitative | In situ | Medium |
| FRET | Molecular proximity | Quantitative | In vivo/vitro | High |
Integrating At5g58784 antibody-derived data with computational modeling:
Data normalization and standardization:
Develop calibration curves using recombinant At5g58784 protein
Normalize protein levels to appropriate housekeeping proteins
Account for technical and biological variability
Transform data appropriately for model integration
Model selection and development:
Choose ordinary differential equation (ODE) models for reaction kinetics
Apply Bayesian networks for inferring regulatory relationships
Use flux balance analysis for metabolic pathway integration
Develop agent-based models for spatial dynamics
Parameter estimation approaches:
Estimate kinetic parameters from time-course experiments
Implement sensitivity analysis to identify critical parameters
Use Bayesian parameter estimation with prior knowledge
Validate model predictions with independent experiments
Multi-scale integration strategies:
Connect protein-level data with transcriptomic responses
Link subcellular localization with tissue-level responses
Integrate temporal dynamics of stress responses
Model feedback between metabolic and signaling networks
Effective modeling requires quantitative antibody-based data with appropriate controls and statistical power. Consider consulting with computational biologists for optimal experimental design that facilitates downstream modeling applications.
Adapting the Fabrack-CAR system described in the literature for plant applications:
System components adaptation:
Modify the CAR construct for plant expression systems
Optimize codon usage for plant expression
Select appropriate plant promoters (constitutive or inducible)
Design plant-compatible selection markers
Meditope-enabled antibody engineering:
Engineer At5g58784 antibodies to contain the meditope-binding pocket
Test binding affinity in plant cellular environments
Validate specificity in plant tissue context
Optimize antibody delivery to plant systems
Application scenarios:
Targeted protein degradation in specific cell types
Conditional protein inactivation during development
Cell type-specific proteomics in complex tissues
Monitoring protein dynamics in response to environmental stimuli
Technical challenges and solutions:
Address plant cell wall barriers for antibody delivery
Develop protoplast-based validation systems
Consider transgenic expression of antibody fragments
Test viral delivery systems for transient expression
This innovative approach would require significant adaptation of the mammalian Fabrack-CAR system for plant biology applications, potentially enabling unprecedented control over protein function in specific plant cell types.
For comprehensive integration of antibody-based data with other omics approaches:
Data collection and standardization:
Generate quantitative protein expression data using validated antibodies
Collect matched samples for transcriptomics and metabolomics
Implement consistent experimental conditions across platforms
Develop standardized metadata annotation
Statistical integration methods:
Apply correlation networks to identify relationships
Use partial least squares regression for latent variable modeling
Implement MOFA+ (Multi-Omics Factor Analysis) for dimension reduction
Consider Bayesian data integration frameworks
Biological pathway contextualization:
Map protein data to known metabolic pathways
Analyze protein-metabolite relationships
Identify transcriptional regulation of At5g58784
Study post-translational modification patterns
Validation strategies:
Test key predictions with targeted experiments
Use orthogonal measurements for critical nodes
Implement time-course analyses to capture dynamics
Apply perturbation studies to validate causal relationships
Multi-omics integration provides a systems-level understanding of At5g58784 function within the broader context of plant metabolic and signaling networks, revealing emergent properties not apparent from single-omics approaches.