PtDi19-7 is upregulated under drought, salt, and ABA treatments. Key findings include:
Drought: 20% PEG 6000 induces PtDi19-7 expression within 1–24 hours .
Salt stress: 200 mmol/L NaCl triggers rapid upregulation (peaking at 3–6 hours) .
ABA signaling: PtDi19-7 overexpression in Arabidopsis enhances sensitivity to ABA and improves drought tolerance .
Transgenic plants: Overexpression of PtDi19-7 in Arabidopsis results in delayed seed germination under ABA and mannitol stress, indicating a role in stress adaptation .
Protein interactions: PtDi19-7 physically interacts with PtDi19-2, forming heterodimers that may amplify stress-responsive gene regulation .
RNA-seq data: PtDi19-7 shows tissue-specific expression, with highest levels in roots under drought stress .
qRT-PCR validation: Confirms rapid induction of PtDi19-7 under ABA treatment (100 µmol/L) .
GFP fusion assays: PtDi19-7 localizes to the nucleus, consistent with its role as a transcription factor .
6. Implications for Future Research
While DI19-7 itself is not an antibody, its study provides insights into stress adaptation mechanisms in plants. Future directions include:
Engineering crops with enhanced DI19-7 expression for drought resilience.
Investigating cross-species functional conservation in agriculturally relevant plants.
DI19-7 Antibody is a research reagent developed to target the Protein DEHYDRATION-INDUCED 19 homolog 7 (DI19-7) in Arabidopsis thaliana. This antibody recognizes specific epitopes on the DI19-7 protein, which plays a role in plant dehydration response pathways . The antibody is primarily used in experimental contexts such as ELISA and Western Blot applications for the identification and quantification of DI19-7 protein in plant samples. Understanding the target specificity is crucial when designing experiments, as it determines the utility and limitations of the antibody in different research applications.
Proper storage and handling of DI19-7 Antibody is critical to maintaining its reactivity and specificity. Generally, antibodies should be stored according to manufacturer recommendations, which typically include keeping them at -20°C for long-term storage or at 4°C for short-term use (1-2 weeks). Repeated freeze-thaw cycles should be avoided as they can lead to protein denaturation and loss of antibody activity. When handling, minimize exposure to room temperature, avoid contamination, and use sterile technique. Consider aliquoting the antibody into single-use volumes to prevent repeated freeze-thaw cycles. Additionally, some antibodies require protease inhibitors or stabilizing proteins like BSA in their storage buffer to maintain functionality .
DI19-7 Antibody has been validated for specific laboratory techniques including ELISA and Western Blot (WB) applications for the identification of the antigen . Based on general antibody validation principles, this validation would have involved confirming that the antibody specifically recognizes DI19-7 protein with minimal cross-reactivity to other proteins. When considering using this antibody for applications beyond those explicitly validated, researchers should conduct preliminary validation experiments to ensure suitability for their specific experimental system. Complete validation typically includes assessment of specificity, sensitivity, and reproducibility across different experimental conditions .
Determining the optimal concentration of DI19-7 Antibody requires a systematic titration approach. Start by preparing a series of dilutions of the antibody. If the manufacturer provides a recommended volume (e.g., 5 μL for 1 million cells), use this as your starting point and test serial dilutions both above and below this concentration (e.g., 10 μL, 5 μL, 2.5 μL, 1.25 μL) . If no recommendations are available, begin with approximately 0.2 μg of antibody as a starting point, calculating the required volume based on the formulated antibody concentration .
For each concentration, assess both qualitative factors (visual separation of positive and negative populations on flow plots or clarity of bands on Western blots) and quantitative measurements (signal-to-noise ratio, median fluorescence intensity). The optimal concentration should provide clear separation between positive and negative populations with minimal background staining. Remember that optimal concentrations may differ between applications (ELISA, Western blotting, etc.), so titration should be performed for each specific application .
When working with DI19-7 Antibody, several controls are essential for experimental rigor:
Positive Control: Samples known to express DI19-7 protein, such as Arabidopsis tissue under dehydration stress conditions.
Negative Control: Samples known not to express the target protein or tissue from knockout plants lacking DI19-7 expression.
Isotype Control: An antibody of the same isotype but with irrelevant specificity to assess non-specific binding.
Secondary Antibody Only Control: Samples treated with only the secondary detection reagent to identify any background signal.
Blocking Control: Samples where the antibody is pre-incubated with purified antigen to confirm specificity .
These controls help distinguish specific from non-specific signals and validate experimental findings. Additionally, when establishing new protocols, include internal controls for normalization and standardization across experiments.
Confirming the specificity of DI19-7 Antibody requires multiple complementary approaches:
Western Blot Analysis: Verify that the antibody detects a protein of the expected molecular weight for DI19-7. Check for absence of non-specific bands.
Immunoprecipitation followed by Mass Spectrometry: This can identify what proteins are actually being pulled down by the antibody.
Genetic Controls: Use DI19-7 knockout or knockdown specimens alongside wild-type samples to confirm absence of signal in the genetic models.
Pre-absorption Test: Pre-incubate the antibody with purified DI19-7 protein before applying to samples. Specific binding should be substantially reduced.
Cross-reactivity Testing: Test the antibody against related proteins (other DI19 family members) to assess potential cross-reactivity.
Validation across Multiple Techniques: Confirm consistent results across different methods (IHC, IF, ELISA, etc.) that use different sample preparation approaches .
These approaches collectively provide strong evidence for antibody specificity and should be documented in your experimental reports.
High background signal when using DI19-7 Antibody can stem from several sources:
Antibody Concentration: Excessive antibody leads to non-specific binding. Solution: Perform a proper titration experiment to determine optimal concentration. Using an antibody blocking buffer (e.g., BSA) can significantly reduce unwanted binding, as demonstrated in Figure 2 of the referenced quality of reagents document .
Insufficient Blocking: Inadequate blocking allows antibody binding to non-specific sites. Solution: Optimize blocking conditions by testing different blockers (BSA, non-fat milk, normal serum) and concentrations.
Cross-reactivity: The antibody may recognize epitopes on proteins similar to DI19-7. Solution: Increase stringency of washing steps and consider pre-absorbing the antibody with related proteins.
Sample Preparation Issues: Over-fixation can expose hydrophobic regions. Solution: Optimize fixation protocols.
Detection System Sensitivity: Some secondary antibodies or detection reagents can amplify background. Solution: Try different detection systems or reduce detection reagent concentration.
Non-specific Interactions: Electrostatic interactions between antibody and sample components. Solution: Increase salt concentration in wash buffers or add detergents like Tween-20 .
Methodically testing these variables will help identify and address the specific cause of high background in your experimental system.
Inconsistent results with DI19-7 Antibody across experiments may occur due to several factors:
Antibody Degradation: Repeated freeze-thaw cycles or improper storage can degrade antibody activity. Solution: Aliquot antibodies for single use and maintain proper storage temperatures.
Batch-to-Batch Variation: Different lots may have varying properties. Solution: Record lot numbers and validate each new lot against previous successful experiments.
Sample Preparation Variability: Inconsistencies in sample handling. Solution: Standardize sample collection, processing, and storage procedures.
Protocol Deviations: Minor changes in incubation times, temperatures, or buffer compositions. Solution: Create detailed protocols with minimal variation and maintain thorough laboratory records.
Equipment Variation: Different instruments or settings between experiments. Solution: Standardize equipment settings and maintenance schedules.
Reagent Quality: Degraded buffers or detection reagents. Solution: Prepare fresh reagents or validate existing ones before critical experiments.
To address inconsistency, implement a systematic troubleshooting approach by changing one variable at a time and documenting all experimental conditions meticulously. Consider running a control sample across multiple experiments to establish a baseline for comparison .
Weak or absent signal when using DI19-7 Antibody may result from several issues:
Insufficient Antigen: Low expression or degradation of the target protein. Solution: Use positive control samples known to express DI19-7 at detectable levels, and optimize sample preparation to preserve protein integrity.
Antibody Activity Loss: Degraded or denatured antibody. Solution: Test a new aliquot or lot of antibody and verify proper storage conditions.
Epitope Masking: Sample preparation may alter or hide the epitope. Solution: Try different antigen retrieval methods (for IHC/IF) or different sample preparation protocols.
Insufficient Incubation: Inadequate antibody-antigen interaction time. Solution: Optimize incubation time and temperature, potentially using longer incubations at 4°C.
Suboptimal Detection System: Secondary antibody or detection reagent issues. Solution: Verify detection system functionality with a different primary antibody known to work.
Buffer Incompatibility: Certain buffer components may interfere with binding. Solution: Test alternative buffer systems recommended for similar antibodies.
PMT Voltage Settings: For flow cytometry applications, inappropriate voltage settings can make populations appear too dim. Solution: Properly adjust PMT voltage so both negative and positive populations are visible on scale, as shown in Figure 3 of the referenced quality document .
Methodically testing these variables and documenting results will help identify the specific cause of weak signals in your experimental system.
Adapting DI19-7 Antibody for multiplex immunofluorescence requires careful optimization:
Antibody Compatibility: First, determine if the DI19-7 Antibody can be used alongside other antibodies without interference. Test for cross-reactivity between all antibodies in your panel, particularly if they originate from the same species.
Fluorophore Selection: Choose fluorophores with minimal spectral overlap. Consider brightness hierarchy - pair dim fluorophores with abundant targets and bright fluorophores with less abundant targets. The DI19-7 Antibody may be available with different conjugates (FITC, Biotin, etc.) that can be selected based on experimental needs .
Sequential Staining Protocol: If using antibodies from the same species, implement sequential staining with blocking steps between antibody applications.
Proper Controls: Include single-stained samples for compensation and fluorescence-minus-one (FMO) controls to set accurate gates.
Optimization of Individual Antibodies: Titrate each antibody individually before combining them to determine optimal concentrations.
Fixation Compatibility: Ensure your fixation method preserves all epitopes of interest without causing autofluorescence.
Signal Amplification Systems: For low-abundance targets, consider using amplification systems like tyramide signal amplification (TSA).
Methodical validation at each step ensures reliable multiplex detection while minimizing false positives from cross-reactivity or spectral overlap .
For quantitative analysis of protein expression using DI19-7 Antibody, several critical factors must be addressed:
Antibody Linearity Range: Determine the dynamic range within which the antibody response is linear to increasing amounts of target protein. This requires creating a standard curve with known quantities of purified DI19-7 protein.
Standardization: Include internal standards in every experiment for normalization. For Western blots, this could be housekeeping proteins; for flow cytometry, standardized beads.
Technical Replicates: Perform multiple technical replicates to assess experimental variability and increase confidence in quantitative measurements.
Signal Saturation: Ensure detection methods operate within non-saturating conditions. Saturated signals will underestimate differences between samples.
Background Subtraction: Implement consistent background subtraction methodologies across all samples.
Equipment Calibration: Regularly calibrate all measurement equipment (flow cytometers, plate readers, imaging systems) to maintain consistency.
Data Analysis Protocols: Establish standardized data analysis workflows that include appropriate statistical methods for quantitative comparisons.
Validation across Methods: Confirm quantitative findings using orthogonal methods (e.g., ELISA, qPCR, mass spectrometry) to increase confidence in results .
These considerations are essential for generating reproducible and reliable quantitative data when using DI19-7 Antibody in research applications.
The performance comparison between monoclonal and polyclonal versions of DI19-7 Antibody involves several key considerations:
Monoclonal DI19-7 Antibody:
Specificity: Typically offers higher specificity by recognizing a single epitope, reducing cross-reactivity with similar proteins.
Consistency: Produces more consistent lot-to-lot performance due to derivation from a single B-cell clone.
Application Strength: Often preferable for applications requiring high specificity such as therapeutic development, flow cytometry, and quantitative assays.
Limitations: May be more sensitive to epitope modifications (e.g., from fixation or denaturation) and might provide weaker signals in some applications.
Polyclonal DI19-7 Antibody:
Epitope Recognition: Recognizes multiple epitopes on DI19-7, potentially providing stronger signals through multiple binding sites.
Robustness: Generally more tolerant to sample preparation variations that might alter protein conformation.
Application Strength: Often preferred for applications like immunoprecipitation and immunohistochemistry, particularly when protein conformation may be altered.
Limitations: Higher potential for cross-reactivity and batch-to-batch variation.
The choice between monoclonal and polyclonal DI19-7 Antibody should be determined by the specific research application, with polyclonal antibodies (such as the rabbit polyclonal mentioned in the search results) often being more versatile across multiple applications (ELISA, IHC, IF) but potentially showing higher background in some contexts .
DI19-7 Antibody can be strategically employed in plant stress response research through several methodological approaches:
Temporal Expression Analysis: Use the antibody to track DI19-7 protein levels at different time points following exposure to drought, salinity, or other stressors. This temporal profiling can reveal the kinetics of stress response activation in Arabidopsis thaliana.
Tissue-Specific Expression Studies: Employ immunohistochemistry with DI19-7 Antibody to map protein expression across different plant tissues under stress conditions, helping identify which tissues prioritize this response pathway.
Protein-Protein Interaction Studies: Utilize co-immunoprecipitation with DI19-7 Antibody to identify interaction partners that change during stress conditions, providing insights into stress response signaling networks.
Subcellular Localization Tracking: Use immunofluorescence microscopy with DI19-7 Antibody to monitor potential changes in protein localization during stress responses, as many transcription factors translocate under stress conditions.
Comparative Analysis Across Ecotypes: Apply the antibody to study DI19-7 expression in different Arabidopsis ecotypes with varying drought tolerance, potentially correlating protein levels with stress resilience phenotypes.
Signal Pathway Activation: Use phospho-specific versions of the antibody (if available) to determine activation states of the protein during stress response.
Transgenic Validation Studies: Employ the antibody to verify protein expression levels in DI19-7 overexpression or knockout lines used in functional studies .
These approaches can generate comprehensive understanding of how DI19-7 contributes to plant adaptive responses to environmental stressors.
When employing DI19-7 Antibody for cross-species studies of conserved dehydration response mechanisms, several methodological considerations are essential:
Epitope Conservation Analysis: Before experimental work, conduct bioinformatic analysis of sequence homology in the epitope region recognized by the antibody across target species. Higher conservation suggests better cross-reactivity potential.
Stepwise Validation Approach: Begin with closely related species (other Brassicaceae family members) before attempting detection in more distantly related plants. Document cross-reactivity patterns systematically.
Western Blot Validation: For each new species, first validate antibody specificity via Western blot, looking for bands of the expected molecular weight based on sequence homology predictions.
Positive Control Inclusion: Always run samples from Arabidopsis thaliana (the original target species) alongside experimental samples as positive controls.
Sensitivity Adjustments: Cross-species applications may require higher antibody concentrations or modified detection methods to account for reduced binding affinity.
Preabsorption Controls: Perform preabsorption controls with purified Arabidopsis DI19-7 protein to confirm specificity of cross-species detection.
Complementary Methods: Supplement antibody-based detection with nucleic acid-based methods (qPCR) to correlate protein detection with gene expression patterns.
Custom Antibody Consideration: For distantly related species, consider developing custom antibodies against conserved regions of the homologous proteins .
These methodological approaches ensure reliable cross-species analysis while minimizing false positives from non-specific binding or false negatives from insufficient epitope recognition.
Advances in antibody engineering are poised to significantly enhance future versions of DI19-7 Antibody and similar research reagents through several innovative approaches:
Improved Specificity Engineering: Techniques like complementarity-determining region (CDR) optimization could produce DI19-7 antibodies with enhanced specificity, reducing cross-reactivity with other DI19 family members while maintaining high affinity for the target.
Multi-epitope Recognition: Development of bispecific or multispecific antibodies that can simultaneously bind multiple epitopes on DI19-7 or recognize both DI19-7 and functionally related proteins, enabling more complex pathway analysis in single experiments.
Environmentally Responsive Modifications: Engineering antibodies with fluorescent reporters that change properties upon binding or in response to environmental conditions (pH, redox state), allowing real-time monitoring of not just protein presence but also its microenvironment.
Enhanced Stability Formats: Creation of single-chain variable fragments (scFvs) or nanobodies derived from DI19-7 antibodies that maintain specificity while offering superior tissue penetration and stability under harsh experimental conditions.
Site-specific Conjugation Methods: Development of site-specific conjugation techniques that preserve antibody function while allowing precise control over the location and number of conjugated molecules (fluorophores, enzymes, etc.).
Recombinant Production Platforms: Transitioning from hybridoma-based to recombinant production methods, similar to what was seen with the human neutralizing antibody P36-5D2 , ensuring greater batch-to-batch consistency and eliminating dependence on animal immunization.
In vitro Selection Methods: Application of display technologies (phage, yeast, or ribosome display) to rapidly generate and screen antibody variants with optimized properties for specific applications .
These engineering advances would significantly expand the utility of DI19-7 antibodies in both basic research and potential biotechnology applications while improving reproducibility across research groups.