ASPGB1 participates in nitrogen recycling, particularly during seed germination and stress responses. It works alongside asparagine synthetase (ASN) to regulate asparagine pools, which are critical for amino acid transport and storage .
Tissue specificity: Highly expressed in vascular tissues and developing seeds .
Regulation: Expression is modulated by nitrogen availability and light conditions .
Parameter | Wild-Type (Col-0) | asn3-1 Mutant | Significance |
---|---|---|---|
Asparagine (Asn) | 100% | 58% | p < 0.01 |
Glutamine (Gln) | 100% | 101% | Not significant |
ASPGB1 mRNA Levels | 100% | 22% | p < 0.001 |
Immunogen: Synthetic peptide from ASPGB1 C-terminal region .
Cross-reactivity: Specific to Arabidopsis thaliana; no reactivity reported in maize or rice .
Knockout mutants (asn3-1): Exhibit 42% reduction in free asparagine levels, impairing seed germination under low-nitrogen conditions .
Stress responses: ASPGB1 expression increases during sulfur deprivation, suggesting a role in stress-induced nitrogen remobilization .
Feature | ASPGB1 (At3g16150) | ASPGA1 (At5g08100) |
---|---|---|
Subcellular Localization | Cytosol | Vacuole |
Substrate Preference | L-asparagine | β-L-Asp-L-Ala |
Expression Pattern | Seed-specific | Ubiquitous |
AT3G16150 encodes a transcript in Arabidopsis thaliana that has been identified as a target of the endoribonuclease DNE1. This gene is particularly significant because it exhibits a dual targeting pattern, showing both decreased and increased 5' monophosphate mRNA fragments (5'P) at different positions when DNE1 is mutated . This dual targeting pattern suggests a complex regulatory mechanism involving both DNE1-mediated cleavage and mRNA decapping. Understanding AT3G16150 regulation provides insights into post-transcriptional control mechanisms in plants, particularly the coordination between endoribonuclease activity and the decapping machinery.
For robust AT3G16150 antibody validation, implement a multi-faceted approach:
Western blot analysis comparing wild-type plants with at3g16150 knockout mutants
Immunoprecipitation followed by mass spectrometry verification
Immunofluorescence microscopy comparing antibody signal in wild-type versus mutant tissues
Preabsorption tests with recombinant AT3G16150 protein
Cross-reactivity assessment against closely related proteins
Testing antibody specificity in diverse experimental conditions is crucial, as binding properties may vary depending on native protein conformation, post-translational modifications, and cellular compartmentalization. Document lot-to-lot variation if using polyclonal antibodies to ensure reproducibility across experiments.
To investigate AT3G16150 interactions with DNE1, consider these complementary approaches:
Technique | Application | Advantages | Limitations |
---|---|---|---|
HyperTRIBE | Identifies direct RNA-protein contacts in vivo | Captures transient interactions; works in native context | Doesn't confirm cleavage events |
GMUCT | Maps 5'P RNA degradation intermediates | Identifies actual cleavage products | Only detects stable degradation products |
Co-immunoprecipitation | Validates protein-protein interactions | Identifies protein complexes | May not distinguish direct from indirect interactions |
RNA-immunoprecipitation | Confirms RNA-protein binding | Works with native complexes | Cannot pinpoint exact binding sites |
The combination of HyperTRIBE and GMUCT has proven particularly effective, as demonstrated in recent research that identified AT3G16150 as both contacting DNE1 and being processed by this endoribonuclease . This dual approach overcomes the limitations of each individual method.
Epitope mapping is essential for characterizing different AT3G16150 antibodies. Implement the following protocol:
Generate a series of truncated AT3G16150 recombinant proteins or peptide arrays covering the entire sequence
Perform western blot analysis with different antibodies against these fragments
Use competitive binding assays with synthetic peptides corresponding to predicted epitopes
Consider phage display techniques for fine-mapping of conformational epitopes
Validate findings with mutational analysis of key residues within identified epitopes
Different epitopes may be accessible in different experimental contexts. For instance, antibodies targeting regions involved in protein-protein interactions might show reduced binding in native complexes but strong reactivity in denatured samples. This information is crucial when selecting antibodies for specific applications such as immunoprecipitation versus western blotting.
For successful AT3G16150 immunoprecipitation from plant tissues:
Sample preparation:
Harvest tissue in optimal physiological state (consider diurnal regulation)
Flash-freeze in liquid nitrogen and grind to fine powder
Extract in buffer containing 50 mM Tris-HCl (pH 7.5), 150 mM NaCl, 5 mM EDTA, 0.1% Triton X-100, 10% glycerol, with freshly added protease inhibitors
Antibody coupling:
Pre-clear lysate with Protein A/G beads (1 hour, 4°C)
Incubate antibody with Protein A/G beads (2 hours, 4°C) or consider covalent coupling using dimethyl pimelimidate
Use 2-5 μg antibody per mg of total protein
Immunoprecipitation:
Incubate pre-cleared lysate with antibody-coupled beads (overnight, 4°C with gentle rotation)
Wash 4-5 times with decreasing salt concentrations
Elute with gentle conditions to maintain protein interactions or harsher conditions for higher purity
Include appropriate controls, including IgG isotype control and immunoprecipitation from knockout plants, to identify non-specific interactions.
For precise AT3G16150 subcellular localization:
Fixation options:
For general immunofluorescence: 4% paraformaldehyde in PBS (pH 7.4) for 20-30 minutes
For preserving membrane structures: Mixture of 4% paraformaldehyde with 0.1-0.5% glutaraldehyde
For nucleic acid-associated proteins: Consider Carnoy's fixative to better preserve nucleic acid structures
Tissue processing:
Create 5-10 μm sections using a cryostat or microtome
Perform antigen retrieval if necessary (10 mM sodium citrate, pH 6.0, 95°C for 10 minutes)
Block with 5% BSA, 5% normal serum, 0.3% Triton X-100 in PBS (1 hour, room temperature)
Immunostaining:
Primary antibody: Use AT3G16150 antibody at 1:100-1:500 dilution (optimize empirically)
Secondary antibody: Fluorophore-conjugated at 1:500-1:1000 dilution
Include DAPI (1 μg/ml) for nuclear staining
Mount in anti-fade medium to preserve fluorescence
Controls and validation:
Include secondary-only controls
Use tissues from knockout plants as negative controls
Consider co-staining with organelle markers for precise localization
This protocol should be optimized based on tissue type and specific experimental requirements.
To investigate AT3G16150 interactions with decapping components:
Sequential immunoprecipitation approach:
First immunoprecipitation with AT3G16150 antibody
Elution under mild conditions
Second immunoprecipitation with antibodies against decapping components (DCP1, DCP2, etc.)
Analysis of doubly-purified complexes by western blot or mass spectrometry
Proximity ligation assay (PLA):
Use primary antibodies against AT3G16150 and decapping components
Apply species-specific PLA probes with oligonucleotide extensions
Ligase joins oligonucleotides if proteins are in close proximity (<40 nm)
Amplify signal by rolling circle amplification
Quantify interaction signals by fluorescence microscopy
FRET-based immunofluorescence:
Use fluorophore-conjugated antibodies against AT3G16150 and decapping components
Measure fluorescence resonance energy transfer to detect close molecular proximity
Perform acceptor photobleaching to confirm specific FRET signals
These techniques can reveal whether AT3G16150 physically interacts with the decapping machinery, supporting the dual targeting model observed in GMUCT data .
For phospho-specific AT3G16150 antibodies:
Phosphorylation site identification:
Perform phosphoproteomic analysis of AT3G16150 immunoprecipitated from plants
Identify conserved phosphorylation motifs using bioinformatics
Prioritize sites showing differential phosphorylation under relevant conditions
Phosphopeptide design:
Synthesize 10-15 amino acid peptides containing the phosphorylated residue centrally positioned
Include terminal cysteine for carrier protein conjugation
Prepare both phosphorylated and non-phosphorylated versions of each peptide
Immunization and screening strategy:
Immunize rabbits with phosphopeptide conjugated to KLH
Screen serum by ELISA against both phosphorylated and non-phosphorylated peptides
Perform dual-affinity purification:
a. Positive selection on phosphopeptide column
b. Negative selection on non-phosphopeptide column
Validation in plant systems:
Test antibody specificity using phosphatase-treated samples
Validate with phosphomimetic and phospho-dead AT3G16150 mutants
Confirm specificity with CRISPR-edited plants where phosphorylation sites are mutated
This meticulous approach will generate highly specific reagents for studying the functional significance of AT3G16150 phosphorylation in DNE1-mediated RNA degradation pathways.
Integrating native mass spectrometry with immunopurification:
Gentle complex isolation:
Extract plant tissue in non-denaturing buffer (e.g., 150 mM ammonium acetate, pH 7.5)
Perform immunoprecipitation with AT3G16150 antibodies conjugated to magnetic beads
Use competitive elution with excess epitope peptide to maintain complex integrity
Native MS sample preparation:
Buffer exchange into MS-compatible volatile buffer (ammonium acetate)
Remove detergents using specialized columns or phase separation techniques
Concentrate sample to 1-5 μM using centrifugal filters with appropriate MWCO
Native MS analysis:
Use nanoelectrospray ionization (nESI) with gentle ionization parameters
Optimize instrument parameters to preserve non-covalent interactions
Include collision-induced dissociation (CID) experiments at varying energies
Data analysis workflow:
Identify stoichiometry of AT3G16150-containing complexes
Characterize complex architecture through partial disruption
Detect conformational changes in response to physiological stimuli
This approach can reveal how AT3G16150 participates in dynamic complexes with RNA degradation machinery components, potentially showing condition-dependent interactions with DNE1 and decapping proteins that would explain the dual targeting patterns observed in degradome studies .
To overcome non-specific binding:
Optimization of blocking conditions:
Test different blocking agents: 5% non-fat milk, 5% BSA, commercial blocking reagents
Evaluate various detergents (Tween-20, Triton X-100) at different concentrations
Consider adding 0.1-0.2% SDS to reduce hydrophobic interactions
Antibody dilution matrix:
Test primary antibody at dilutions from 1:100 to 1:10,000
Combine with secondary antibody dilutions from 1:1,000 to 1:20,000
Assess signal-to-noise ratio at each combination
Sample preparation modifications:
Extend pre-clearing steps with appropriate beads
Include competing proteins (e.g., 0.1-0.5 mg/ml sheared salmon sperm DNA for nucleic acid-binding proteins)
Consider crosslinking optimization if working with fixed samples
Advanced purification approaches:
Perform subtractive purification against knockout plant extracts
Consider affinity purification against recombinant AT3G16150
Use sequential affinity steps to improve specificity
Document all optimization parameters systematically to identify patterns in non-specific binding and develop a reproducible protocol for high-specificity applications.
To investigate AT3G16150's dual targeting mechanism:
Immunoprecipitation of degradation intermediates:
Use AT3G16150 antibodies to capture full-length and partially degraded forms
Analyze 5' ends of co-purified RNA using modified RACE protocols
Compare profiles between wild-type, dne1, dcp2, and dne1 dcp2 double mutants
In vitro reconstitution system:
Express and purify recombinant AT3G16150, DNE1, and decapping components
Perform sequential or simultaneous incubation experiments
Monitor degradation patterns using labeled RNA and gel electrophoresis
Use antibodies to immunodeplete specific factors
Pulse-chase experimental design:
Generate transgenic plants expressing epitope-tagged AT3G16150 under inducible promoter
Perform transcriptional pulse-chase experiments
Immunoprecipitate AT3G16150 mRNA at different time points
Analyze degradation intermediates by Northern blot or sequencing
Single-molecule approaches:
Label AT3G16150 mRNA with fluorescent probes
Add fluorescently labeled recombinant DNE1 and decapping enzymes
Monitor interaction dynamics using total internal reflection fluorescence microscopy
Use antibodies to verify protein identities in observed complexes
These approaches would provide mechanistic insight into how AT3G16150 mRNA is recognized and processed by both DNE1 and decapping pathways, expanding on the observations from GMUCT data .
To distinguish direct from indirect interactions:
Protein crosslinking strategy:
Apply gradient of crosslinker concentrations (0.1-2% formaldehyde)
Perform AT3G16150 immunoprecipitation followed by western blotting
Compare interaction patterns at different crosslinking strengths
Direct interactions typically persist at lower crosslinker concentrations
In vitro binding assays:
Express recombinant AT3G16150 and potential interacting partners
Perform pull-down assays with purified components
Include appropriate negative controls (unrelated proteins of similar size/charge)
Quantify binding using surface plasmon resonance or bio-layer interferometry
Yeast three-hybrid approach:
Design system with AT3G16150 as bait
Test interactions with DNE1 and decapping components
Include bridge proteins to test for indirect interactions
Compare interaction strength in quantitative β-galactosidase assays
Antibody-based competitive disruption:
Use epitope-specific antibodies targeting different domains of AT3G16150
Add antibodies to in vitro or semi-permeabilized cell systems
Monitor which interactions are disrupted by specific antibodies
Map interaction interfaces through systematic epitope blocking
These methodologies would help create an interaction map for AT3G16150, revealing whether its dual targeting by DNE1 and decapping machinery involves direct physical interactions or is mediated through adapter proteins or RNA structural elements.
Adapting transcriptome-wide methodologies to study epitope accessibility:
Modified HyperTRIBE approach:
Generate fusion proteins with ADAR linked to AT3G16150-interacting proteins
Create parallel constructs with ADAR fused to AT3G16150 antibody epitope tags
Compare editing patterns to identify regions where antibody binding might compete with protein interactions
Use computational modeling to predict epitope masking in different complexes
Antibody-coupled GMUCT strategy:
Perform immunoprecipitation with AT3G16150 antibodies under different conditions
Extract and sequence associated RNA fragments
Compare degradation patterns in immunoprecipitated versus total RNA pools
Identify condition-specific changes in epitope accessibility
Integration with structural biology:
Use hydrogen-deuterium exchange mass spectrometry with and without antibody binding
Apply crosslinking mass spectrometry to map protein interaction surfaces
Develop computational models predicting epitope accessibility based on interaction data
Validate with cryoEM of AT3G16150-containing complexes
This integrated approach would provide unprecedented insights into how AT3G16150 epitope accessibility changes during mRNA processing, potentially explaining why certain antibodies might fail to detect AT3G16150 in specific functional states despite the protein being present.
Advanced antibody engineering strategies:
Single-chain variable fragment (scFv) development:
Clone variable regions from validated AT3G16150 monoclonal antibodies
Express as scFv fusion with subcellular targeting sequences
Monitor real-time localization in living plant cells
Compare accessibility of different epitopes across compartments
pH-sensitive antibody variants:
Introduce histidine substitutions at key positions in CDR loops
Engineer antibodies with differential binding at cytosolic versus vacuolar pH
Apply to track AT3G16150 movement between compartments
Validate with ratiometric imaging approaches
Split-antibody complementation systems:
Design antibody fragments that reassemble when AT3G16150 is in specific compartments
Pair with compartment-specific marker proteins
Generate fluorescent or enzymatic readout upon complementation
Use to detect rare localization events in specific cellular contexts
Nanobody development strategy:
Immunize camelids with purified AT3G16150 protein
Select nanobodies with diverse epitope recognition and binding properties
Engineer for compartment-specific detection using targeting sequences
Validate with super-resolution microscopy techniques
These approaches would transform AT3G16150 antibodies from simple detection reagents into sophisticated tools for studying protein dynamics across cellular compartments during RNA processing events.