The term "ATL" appears in Search Result , referring to Adult T-cell Leukemia (ATL). This study identifies antibodies targeting an antigen in MT-1 cells from ATL patients, but these antibodies are not named "ATL68" . Separately, CD68 antibodies are discussed in Search Results , but "CD68" and "ATL68" are distinct entities.
Antibodies are typically named based on target antigens, clone identifiers, or research contexts (e.g., "VHH PFFNB2" in α-synuclein research ).
The absence of "ATL68" in academic databases (e.g., PubMed, Human Protein Atlas) suggests it is either an obsolete term, a proprietary designation not widely published, or a misinterpretation of combined terms like "ATL" and "CD68."
While "ATL68" remains unverified, key antibody types in ATL and related oncology include:
CD68 Antibodies: These target a glycoprotein marker for macrophages and are validated in Western blot, IHC, and immunofluorescence . No linkage to ATL is documented.
MT-1 Cell Antigen Antibodies: Studied in ATL, these antibodies react with cytoplasmic antigens in MT-1 cells but lack the "ATL68" designation .
To address the query effectively, additional steps would be required:
Clarify Terminology: Verify if "ATL68" refers to a specific clone, commercial product, or preclinical candidate.
Expand Source Review: Search proprietary antibody catalogs (e.g., Thermo Fisher, BioLegend) or niche oncology repositories.
Validate Specificity: If "ATL68" is a novel antibody, experimental data (e.g., ELISA, epitope mapping) would be needed to confirm its target and utility.
ATL68 Antibody is a polyclonal antibody raised in rabbits against recombinant Arabidopsis thaliana ATL68 protein. This antibody specifically recognizes and binds to ATL68 protein in Arabidopsis thaliana (Mouse-ear cress), a model organism widely used in plant biology research . For experimental applications, this antibody has been validated for Enzyme-Linked Immunosorbent Assay (ELISA) and Western Blotting (WB) techniques to detect the target protein in plant tissue samples .
To maintain antibody activity and prevent degradation, ATL68 Antibody should be stored at either -20°C or -80°C immediately upon receipt . Multiple freeze-thaw cycles should be strictly avoided as they can compromise antibody function through protein denaturation. The antibody is supplied in a protective storage buffer containing 0.03% Proclin 300 as a preservative, 50% Glycerol for stability, and 0.01M PBS at pH 7.4 to maintain proper ionic environment . If partial usage is anticipated, aliquoting into smaller volumes before freezing is recommended to minimize freeze-thaw damage.
ATL68 Antibody demonstrates high specificity for its target in Arabidopsis thaliana due to its development using antigen affinity purification methods . Unlike many commercially available plant antibodies that show cross-reactivity across multiple species, the ATL68 Antibody has been specifically tested and optimized for reactivity with Arabidopsis thaliana proteins . This targeted specificity makes it particularly valuable for research focused on precise protein localization and functional studies in this model organism. Similar to antibodies developed through the Recombinant Antibody Network (RAN), this antibody benefits from standardized development processes designed to ensure reproducibility and reliable performance in experimental applications .
The ATL68 Antibody has been specifically validated for ELISA and Western Blot (WB) applications, with recommended dilution ratios of 1:2000 for WB applications . These validated applications enable researchers to:
Detect and quantify ATL68 protein expression levels in plant tissue extracts via ELISA
Determine the molecular weight and expression patterns of ATL68 protein in different tissues or under various experimental conditions using Western Blot
Confirm protein identity through antigen detection in fractionated samples
Unlike antibodies such as the LF-68 antibody which has been validated for immunohistochemistry at 1:400 dilution , the ATL68 Antibody has not been specifically validated for IHC applications, though researchers may optimize protocols for this purpose.
ATL68 Antibody is produced through immunization of rabbits with a recombinant Arabidopsis thaliana ATL68 protein immunogen . Following immunization and antibody production, the antibody undergoes antigen affinity purification to isolate only those antibodies that specifically bind to the target protein, ensuring high specificity . The final product is a polyclonal IgG preparation in liquid form, supplied in a buffer containing preservative and stabilizing agents . This approach differs from monoclonal antibody production methods used for therapeutic antibodies like those described in research by Florida State University chemists, which involve more complex isolation of single antibody-producing cell lines .
ATL68 Antibody provides a valuable tool for investigating plant stress responses and protein regulation mechanisms in Arabidopsis thaliana. Methodologically, researchers can implement:
Time-course experiments: Using the antibody in Western blot analyses to track ATL68 protein levels at different time points after exposure to biotic or abiotic stresses (e.g., pathogen infection, drought, temperature changes)
Subcellular fractionation studies: Combining cell fractionation with immunoblotting to determine the subcellular localization of ATL68 protein under different experimental conditions
Co-immunoprecipitation (Co-IP): Using ATL68 Antibody to pull down the target protein along with its interaction partners to elucidate signaling pathways and regulatory networks
Immunofluorescence microscopy: Optimizing the antibody for microscopy to visualize spatial distribution of ATL68 in plant tissues
Similar to approaches used with other plant antibodies, these methods can reveal valuable insights into protein function and regulation, comparable to how researchers at UCSF Recombinant Antibody Network have used antibodies to understand cell-surface protein dynamics .
When interpreting Western blot results using ATL68 Antibody, researchers should consider several technical and biological factors:
Signal specificity validation: Include appropriate positive and negative controls to confirm signal specificity. Knockout/knockdown lines for ATL68 serve as excellent negative controls to validate antibody specificity
Loading control normalization: Use established plant housekeeping proteins (e.g., actin, tubulin) to normalize protein loading across samples
Technical considerations:
Blocking optimization: Test different blocking agents (e.g., BSA, non-fat milk) to minimize background
Antibody dilution: The recommended 1:2000 dilution may require optimization based on protein abundance
Detection method compatibility: Choose detection systems that provide appropriate sensitivity for your expected protein levels
Data quantification: Use densitometric analysis software to quantify band intensity, applying statistical analysis to determine significance of observed differences
This methodological approach resembles the careful validation processes used in antibody-based research as described in the recent work from Florida State University using advanced imaging techniques for protein analysis .
Protein glycosylation can significantly impact ATL68 Antibody binding efficiency and experimental outcomes. Research on antibody-protein interactions indicates that:
Post-translational modifications (PTMs) like glycosylation can alter epitope accessibility and antibody recognition. Recent studies with NISTmAb demonstrated that while structural variations among antibody populations are not influenced by glycosylation , target protein glycosylation may still affect antibody binding.
Methodological considerations include:
Sample preparation: Treating samples with deglycosylation enzymes before immunodetection can help determine if glycosylation interferes with antibody binding
Protocol modifications: Adjusting detergent types/concentrations or denaturing conditions to improve epitope exposure if glycosylation masks binding sites
Comparative analysis: Running parallel experiments with deglycosylated and native samples to assess differences in detection sensitivity
Data interpretation: Researchers should document molecular weight shifts between predicted and observed protein sizes, as these may indicate the presence of PTMs that could affect antibody binding efficiency.
Similar to the findings reported by Bleiholder's lab using Tandem-TIMS technology to analyze protein structures , these approaches can reveal important insights into how target protein modifications influence experimental outcomes.
When ATL68 Antibody produces unexpected results, systematic troubleshooting approaches are essential:
Problem | Potential Causes | Recommended Solutions |
---|---|---|
No signal | Protein degradation, insufficient antibody concentration, improper detection method | Verify sample integrity with fresh extractions, increase antibody concentration, confirm secondary antibody compatibility |
Multiple bands | Cross-reactivity, protein degradation, post-translational modifications | Use freshly prepared samples with protease inhibitors, optimize blocking conditions, perform peptide competition assay |
High background | Insufficient blocking, excessive antibody concentration, non-specific binding | Extend blocking time, increase blocking agent concentration, titrate primary antibody, increase wash duration and volume |
Inconsistent results | Freeze-thaw cycles, sample preparation variations, buffer incompatibility | Use fresh antibody aliquots, standardize sample preparation protocols, verify buffer compatibility |
Additionally, maintaining detailed laboratory records of experimental conditions and implementing controlled parameter changes will help identify the specific factors affecting antibody performance. This methodical approach aligns with best practices in antibody-based research as evidenced in studies using complex antibody technologies like those developed at the Recombinant Antibody Network .
Integrating ATL68 Antibody immunoprecipitation with mass spectrometry creates powerful opportunities for comprehensive protein characterization:
Immunoprecipitation-Mass Spectrometry (IP-MS) workflow:
Use ATL68 Antibody to selectively isolate the target protein and its interacting partners from plant extracts
Process the immunoprecipitated proteins for mass spectrometry analysis using standard proteomics protocols
Analyze the resulting MS data to identify protein complexes and post-translational modifications
Technical considerations:
Antibody crosslinking to beads prevents antibody contamination in MS samples
Mild elution conditions preserve protein-protein interactions
Tandem mass spectrometry enables identification of specific post-translational modifications
Data integration approach:
Combine Western blot results with MS data to validate protein identifications
Use quantitative MS approaches to determine stoichiometry of protein complexes
Apply bioinformatics tools to map interaction networks centered on ATL68
This combined approach is similar to cutting-edge techniques like Tandem-TIMS (tandem-trapped ion mobility spectrometry) described in research from Florida State University, which allows detailed structural and stability analysis of complex proteins .
Effective sample preparation is critical for successful ATL68 Antibody applications across different plant tissues:
Leaf tissue protocol:
Grind 100-200 mg fresh or frozen leaf tissue in liquid nitrogen to fine powder
Add 500 μl extraction buffer (50 mM Tris-HCl pH 7.5, 150 mM NaCl, 1% Triton X-100, 1 mM EDTA, 1 mM PMSF, protease inhibitor cocktail)
Incubate with gentle agitation for 30 minutes at 4°C
Centrifuge at 14,000 × g for 15 minutes at 4°C
Collect supernatant and determine protein concentration
Add sample buffer and heat at 95°C for 5 minutes for Western blotting applications
Root tissue protocol:
Requires additional washing steps to remove soil contaminants
Includes higher detergent concentration (1.5% Triton X-100) for improved membrane protein extraction
Seed tissue protocol:
Needs extended grinding time due to tissue hardness
Benefits from overnight protein extraction at 4°C to improve yield
Storage recommendations:
These protocols incorporate best practices from plant protein research and are designed to maintain protein integrity while maximizing extraction efficiency.
Proper experimental controls are essential for rigorous validation of ATL68 Antibody performance:
Positive controls:
Recombinant ATL68 protein expression system (e.g., E. coli or plant-based)
Wild-type Arabidopsis thaliana tissues with verified ATL68 expression
Overexpression lines with ATL68 under a strong promoter
Negative controls:
Procedural controls:
Secondary antibody-only control to detect non-specific binding
Loading controls using established housekeeping proteins
Peptide competition assay using immunizing peptide
Application-specific controls:
This comprehensive control strategy mirrors approaches used in advanced antibody validation studies, such as those employed by Atlas Antibodies in their antibody validation workflows .
Optimizing ATL68 Antibody performance under challenging experimental conditions requires systematic protocol adjustments:
For low abundance targets:
Increase protein loading (up to 50-100 μg per lane for Western blotting)
Enhance detection using signal amplification systems (e.g., biotin-streptavidin)
Extend primary antibody incubation to overnight at 4°C
Consider protein enrichment through immunoprecipitation before analysis
For tissues with high proteolytic activity:
Implement stronger protease inhibitor cocktails
Conduct all extraction steps at 4°C
Add reducing agents (e.g., DTT) to maintain protein conformational integrity
Consider immediate sample processing or snap-freezing in liquid nitrogen
For membrane-associated protein targets:
Test different detergents (CHAPS, NP-40, or digitonin) for gentler extraction
Optimize solubilization time and temperature
Explore specialized membrane protein extraction kits
For problematic background:
Test alternative blocking agents (5% BSA often performs better than milk for plant samples)
Increase blocking time and wash duration
Apply gradient washing (increasing stringency with each wash)
Consider using specialized low-background detection systems
These optimization approaches are based on established principles in antibody-based research and reflect methodologies used in advanced antibody applications described in recent literature .
Adapting ATL68 Antibody for co-immunoprecipitation (Co-IP) studies requires specific methodological considerations:
Sample preparation optimization:
Use gentle lysis buffers (e.g., 50 mM Tris-HCl pH 7.5, 150 mM NaCl, 0.5% NP-40, 1 mM EDTA) to preserve protein-protein interactions
Include phosphatase inhibitors to maintain phosphorylation-dependent interactions
Perform protein extraction at 4°C throughout to minimize complex dissociation
Antibody coupling strategy:
Immunoprecipitation protocol:
Pre-clear lysates with protein A/G beads to reduce non-specific binding
Incubate cleared lysates with antibody-coupled beads overnight at 4°C with gentle rotation
Perform stringent washes while maintaining complex integrity
Elute proteins using either gentle (non-denaturing) or harsh (denaturing) conditions depending on downstream applications
Validation and analysis approaches:
Confirm pull-down efficiency by Western blotting for ATL68 protein
Identify interacting partners through mass spectrometry analysis
Validate key interactions through reciprocal Co-IP or other interaction assays
This methodology leverages techniques similar to those used by research groups in the Recombinant Antibody Network for studying protein complexes and interactions .
Using ATL68 Antibody across different plant developmental stages requires several important considerations:
Developmental expression profile:
ATL68 protein levels may vary significantly across developmental stages
Conduct preliminary experiments to establish baseline expression patterns
Adjust antibody concentrations or exposure times accordingly for different stages
Tissue-specific extraction modifications:
Seedling tissues: Require gentler extraction conditions due to delicate nature
Mature tissues: May need more robust extraction methods to overcome structural barriers
Reproductive tissues: Often contain specialized compounds requiring additional purification steps
Stage-specific controls:
Include tissue-matched controls from wild-type plants
Consider using developmentally regulated reference proteins as comparative controls
Document tissue-specific antibody performance characteristics
Quantitative analysis approach:
Normalize ATL68 protein levels to total protein or stage-appropriate housekeeping proteins
Account for developmental variations in protein extraction efficiency
Apply statistical methods appropriate for developmental time-series data
This developmental approach to antibody utilization aligns with best practices in plant molecular biology research and ensures optimal detection across the plant life cycle.
Rigorous quantification and statistical analysis of Western blot data generated with ATL68 Antibody requires a methodical approach:
Image acquisition protocol:
Capture images using a digital imaging system with linear dynamic range
Avoid saturation by optimizing exposure settings
Include internal standards for normalization across blots
Document all image acquisition parameters
Densitometric analysis methodology:
Use specialized software (e.g., ImageJ, Image Lab) for band intensity measurement
Define regions of interest consistently across all samples
Subtract local background from each measurement
Normalize target protein (ATL68) to loading control
Statistical analysis framework:
For comparing two conditions: Apply paired t-test or Wilcoxon signed-rank test based on data distribution
For multiple conditions: Use ANOVA followed by appropriate post-hoc tests (e.g., Tukey's HSD)
For time-course experiments: Consider repeated measures ANOVA or mixed-effects models
Report effect sizes alongside p-values for comprehensive interpretation
Data presentation standards:
Present representative blot images alongside quantification graphs
Include all replicates in statistical analyses
Report sample sizes, statistical tests, and p-values
Use error bars representing standard deviation or standard error as appropriate
This approach to quantitative Western blot analysis ensures robust and reproducible findings, following principles similar to those employed in advanced protein analysis methods described in recent literature .
Addressing non-specific binding with ATL68 Antibody requires systematic troubleshooting strategies:
Optimizing blocking conditions:
Test different blocking agents (BSA, non-fat milk, commercial blocking buffers)
Increase blocking duration from standard 1 hour to 2-3 hours or overnight at 4°C
Evaluate different blocking buffer formulations by adding 0.1-0.3% Tween-20 or Triton X-100
Antibody dilution optimization:
Washing protocol enhancement:
Increase number of wash steps (5-6 washes instead of standard 3)
Extend wash duration from 5 to 10-15 minutes per wash
Use graduated washing stringency (increasing detergent in initial washes, decreasing in later washes)
Sample preparation refinement:
Include additional pre-clearing steps (e.g., pre-incubation with beads)
Test different lysis buffer compositions to reduce interfering compounds
Consider sample pre-treatment with additives that reduce non-specific interactions
Validation strategies:
Conduct peptide competition assays using the immunizing peptide
Compare signal patterns between wild-type and knockout/knockdown samples
Evaluate cross-reactivity using heterologous expression systems
These methodological refinements align with best practices in antibody-based research and reflect approaches used for optimizing antibody performance in complex biological samples .
Differentiating between ATL68 isoforms or post-translationally modified variants requires sophisticated experimental approaches:
Electrophoretic separation strategies:
Use gradient gels (4-15%) to achieve better resolution of closely migrating protein forms
Implement Phos-tag acrylamide gels to separate phosphorylated from non-phosphorylated forms
Apply 2D electrophoresis (isoelectric focusing followed by SDS-PAGE) to resolve isoforms with similar molecular weights but different charge properties
Specialized detection methods:
Employ phospho-specific staining (Pro-Q Diamond) alongside ATL68 Antibody detection
Use modification-specific antibodies in parallel with ATL68 Antibody
Implement sequential reprobing strategies to identify multiple forms on the same membrane
Biochemical discrimination approaches:
Treat samples with specific enzymes (phosphatases, glycosidases, etc.) to remove modifications
Compare migration patterns before and after enzymatic treatment
Use fractional precipitation techniques to enrich specific protein variants
Mass spectrometry integration:
Combine immunoprecipitation with mass spectrometry analysis
Apply targeted MS approaches to identify specific modifications
Quantify relative abundance of different proteoforms
This multi-faceted approach to proteoform analysis draws on advanced protein characterization techniques similar to those used in the innovative imaging research conducted at Florida State University, where technologies like Tandem-TIMS have been applied to study protein structural variations .
Effective membrane stripping and reprobing protocols for ATL68 Antibody require balancing between complete antibody removal and protein retention:
Mild stripping buffer protocol:
Buffer composition: 15 mM glycine, 1% SDS, 1% Tween-20, pH 2.2
Procedure:
Wash membrane in TBST for 5 minutes
Incubate membrane in mild stripping buffer for 10 minutes at room temperature
Repeat incubation with fresh stripping buffer
Wash thoroughly with TBST (3 × 10 minutes)
Re-block membrane before reprobing
Best for: Sequential detection of proteins with substantial size differences
Moderate stripping buffer protocol:
Buffer composition: 62.5 mM Tris-HCl pH 6.8, 2% SDS, 100 mM β-mercaptoethanol
Procedure:
Incubate membrane in buffer for 15-30 minutes at 50°C with gentle agitation
Wash extensively with TBST (6 × 10 minutes)
Verify complete stripping by exposing membrane to detection reagent
Re-block membrane before reprobing
Best for: Most standard reprobing applications with ATL68 Antibody
Strong stripping buffer protocol:
Buffer composition: 62.5 mM Tris-HCl pH 6.7, 2% SDS, 100 mM β-mercaptoethanol, 7 M guanidine hydrochloride
Procedure:
Incubate membrane in buffer for 15 minutes at room temperature
Wash extensively with TBST (6 × 10 minutes)
Verify complete stripping by exposing membrane to detection reagent
Re-block membrane before reprobing
Best for: Removing strongly bound antibodies when other methods fail
Critical considerations:
Document membrane image before stripping as reference
Limit number of stripping cycles (typically 2-3 maximum)
Validate signal reduction after each stripping cycle
Optimize stripping conditions for specific application needs
These protocols provide researchers with options ranging from gentle to aggressive stripping conditions, enabling strategic experimental design based on specific research requirements.
ATL68 Antibody provides valuable tools for investigating protein degradation pathways in plants through several methodological approaches:
Protein stability assessment protocol:
Treat plant tissues or cells with protein synthesis inhibitors (e.g., cycloheximide)
Collect samples at defined time points (0, 1, 3, 6, 12, 24 hours)
Process samples for Western blotting with ATL68 Antibody
Quantify protein levels to determine half-life and degradation kinetics
Proteasome-dependent degradation analysis:
Pre-treat samples with proteasome inhibitors (MG132, bortezomib)
Compare ATL68 protein accumulation with and without inhibitor treatment
Detect ubiquitinated forms using co-immunoprecipitation followed by ubiquitin blotting
Analyze changes in degradation patterns under different stress conditions
Autophagy pathway investigation:
Apply autophagy inhibitors (3-methyladenine, bafilomycin A1)
Track ATL68 protein accumulation in autophagy-deficient mutants
Perform co-localization studies with autophagy markers
Analyze ATL68 fragments that may indicate partial degradation
Experimental design for degradation pathway mapping:
Systematic application of different pathway inhibitors
Genetic manipulation of key degradation pathway components
Stress treatments known to activate specific degradation pathways
Quantitative analysis of degradation rates under different conditions
Treatment | Purpose | Sampling Timepoints | Expected Outcome if ATL68 is Degraded via Pathway |
---|---|---|---|
Cycloheximide | Block protein synthesis | 0, 1, 2, 4, 8 hours | Progressive decrease in ATL68 signal |
MG132 | Block proteasome | 0, 3, 6, 12 hours | Stabilization of ATL68 if proteasome-degraded |
3-MA | Block autophagy | 0, 6, 12, 24 hours | Stabilization of ATL68 if autophagy-degraded |
Combined inhibitors | Pathway discrimination | 0, 6, 12, 24 hours | Reveal primary and secondary pathways |
These methodological approaches enable researchers to decipher the regulatory mechanisms controlling ATL68 protein turnover, similar to strategies used in studying protein dynamics in other biological systems .
Several emerging technologies hold promise for enhancing ATL68 Antibody applications in future plant research:
Advanced imaging technologies:
Super-resolution microscopy allowing visualization of protein distribution with nanometer precision
Expansion microscopy for improved spatial resolution in plant tissues
Correlative light and electron microscopy (CLEM) to connect protein localization with ultrastructural features
Live-cell imaging using antibody fragments for dynamic protein tracking
Single-cell approaches:
Single-cell proteomics to analyze ATL68 levels in individual plant cells
Spatial proteomics for tissue-specific protein mapping
Antibody-based flow cytometry for plant protoplasts
In situ proximity ligation assays for visualizing protein interactions at cellular resolution
High-throughput screening platforms:
Antibody arrays for parallel analysis of multiple proteins
Automated immunoassay systems for large-scale phenotyping
Microfluidic devices for reduced sample requirements
Cell-free expression systems for rapid protein interaction testing
Computational and bioinformatic tools:
Machine learning algorithms for improved image analysis and pattern recognition
Integrative multi-omics approaches combining antibody-based data with transcriptomics and metabolomics
Systems biology modeling of protein networks
Virtual screening tools to predict protein-protein interactions
These technological advances parallel developments in other fields of antibody research, such as the innovative imaging techniques being applied to therapeutic antibodies and disease-related proteins , and represent exciting future directions for plant molecular biology research using ATL68 Antibody.
Integrating ATL68 Antibody-based approaches with genetic manipulation techniques creates powerful research strategies:
CRISPR/Cas9 system integration:
Generate precise ATL68 gene modifications (knockouts, point mutations)
Create epitope-tagged versions at endogenous loci
Validate antibody specificity using CRISPR-generated knockouts
Study protein function through structure-function analysis combining mutations with antibody detection
Transgenic approaches:
Develop inducible expression systems for controlled ATL68 overexpression
Create fluorescent protein fusions for co-localization studies with antibody staining
Implement tissue-specific promoters to study ATL68 function in defined cell types
Generate truncation variants to map antibody epitopes and functional domains
RNA interference and antisense strategies:
Establish knockdown lines with reduced ATL68 expression
Quantify knockdown efficiency using ATL68 Antibody
Compare phenotypic effects of knockdown versus knockout
Create conditional silencing systems for temporal studies
Experimental design considerations:
Include multiple independent transformation lines
Implement proper genetic background controls
Quantify protein levels across transgenic lines using standardized antibody-based assays
Apply statistical methods appropriate for combined genetic-biochemical data
This integrated approach leverages the complementary strengths of antibody-based protein detection and genetic manipulation, similar to strategies employed in advanced research platforms such as those developed by the Recombinant Antibody Network .