DOF5.8 Antibody

Shipped with Ice Packs
In Stock

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
DOF5.8 antibody; At5g66940 antibody; K8A10.1Dof zinc finger protein DOF5.8 antibody; AtDOF5.8 antibody
Target Names
DOF5.8
Uniprot No.

Target Background

Function
DOF5.8 is a transcription factor that specifically binds to the 5'-AA[AG]G-3' consensus core sequence.
Gene References Into Functions
PMID: 25794540, Overexpression of Dof5.8 in *Arabidopsis thaliana* modulates auxin response and impairs vein formation., .
PMID: 25336688, MONOPTEROS (MP) directly activates the Dof5.8 promoter. Analysis of *mp dof5.8* double mutants revealed that Dof5.8 mutations enhance the phenotype of a weak *mp* allele., .
PMID: 25572919, Under abiotic stress, ATDOF5.8 regulates the expression of ANAC069. Activated ANAC069 then binds to NAC recognition sequences or other motifs to regulate the expression of genes containing these motifs in their promoters., .
Database Links

KEGG: ath:AT5G66940

STRING: 3702.AT5G66940.1

UniGene: At.66743

Subcellular Location
Nucleus.

Q&A

What is DOF5.8 and why is it important to detect it in plant research?

DOF5.8 belongs to the DNA binding with One Finger (Dof) family of transcription factors that control plant growth and development, particularly vascular tissue formation. It is expressed early in provascular cells and functions as a transcriptional repressor that modulates auxin response. Its importance lies in its role regulating the formation of higher-order veins in cotyledons and leaves. When overexpressed, DOF5.8 inhibits vein formation, likely through downregulation of auxin-associated transcription factor genes like DORNRöSCHEN and SHI-RELATED SEQUENCE 5 . Detecting DOF5.8 protein levels and localization is crucial for understanding the molecular mechanisms governing vascular patterning in plants.

How can I validate the specificity of a custom DOF5.8 antibody?

Validating antibody specificity for DOF5.8 requires multiple complementary approaches:

  • Use Western blot analysis comparing:

    • Wild-type plant extracts (positive control)

    • dof5.8 mutant extracts (negative control)

    • DOF5.8 overexpression lines (enhanced signal)

  • Perform peptide competition assays where the antibody is pre-incubated with the immunizing peptide before use (specific signal should be abolished)

  • Test cross-reactivity with other Dof family members (DOF4.7, DOF5.1, DOF5.6) which share sequence homology with DOF5.8

  • Include recombinant DOF5.8 protein as a positive control and size reference

  • Validate through immunoprecipitation followed by mass spectrometry to confirm the identity of captured proteins

What tissues and developmental stages are optimal for DOF5.8 detection?

Based on DOF5.8's known expression pattern and function:

  • Developing leaf primordia show highest expression levels during vascular differentiation

  • Young cotyledons during early vascular pattern formation

  • Shoot apical meristems where new leaf primordia emerge

  • Provascular cells in developing organs

The optimal developmental window coincides with early vascular differentiation, as DOF5.8 is expressed in provascular cells under the control of MONOPTEROS/ARF5 . For best results, collect tissues at these early developmental stages and process immediately to prevent protein degradation. Cell-type specific approaches may be necessary as DOF5.8 expression is limited to specific cell populations during development .

What protein extraction methods work best for DOF5.8 detection in plant tissues?

For optimal DOF5.8 extraction from plant tissues:

  • Nuclear extraction methods are recommended as DOF5.8 is a nuclear-localized transcription factor:

    • Use buffers containing 20-50mM Tris-HCl (pH 7.5-8.0), 150mM NaCl, 5mM EDTA, with 0.1-1% detergent (Triton X-100 or NP-40)

    • Include protease inhibitors to prevent degradation

    • Add phosphatase inhibitors if detecting phosphorylation states

  • Sample processing considerations:

    • Flash-freeze tissue samples in liquid nitrogen immediately after collection

    • Grind thoroughly to fine powder while maintaining frozen state

    • Maintain cold temperatures throughout extraction process

    • Clarify extracts by centrifugation at high speed (>12,000g)

  • Enrichment approaches:

    • Nuclear fractionation prior to immunoprecipitation can increase signal-to-noise ratio

    • Density gradient centrifugation may further purify nuclear fractions

How can I optimize conditions for DOF5.8 ChIP (Chromatin Immunoprecipitation) experiments?

Optimizing DOF5.8 ChIP requires careful consideration of several parameters:

  • Crosslinking optimization:

    • Test formaldehyde concentrations (1-3%)

    • Optimize crosslinking time (10-20 minutes at room temperature)

    • Consider dual crosslinkers for improved protein-DNA fixation

  • Chromatin fragmentation:

    • Optimize sonication parameters to achieve 200-500bp fragments

    • Verify fragmentation efficiency by agarose gel electrophoresis

    • Consider micrococcal nuclease digestion as an alternative approach

  • Immunoprecipitation conditions:

    • Antibody concentration (typically 2-5μg per reaction)

    • Incubation time and temperature (overnight at 4°C)

    • Appropriate negative controls (pre-immune serum, IgG)

  • Washing stringency:

    • Optimize salt concentration in wash buffers

    • Include detergent-containing washes to reduce background

    • Test multiple wash protocols to balance signal retention and specificity

  • Positive control regions:

    • Include genomic regions known to be bound by DOF transcription factors

    • Target regions associated with genes downregulated in DOF5.8 overexpressors

What approaches can reveal DOF5.8 protein-protein interactions in vivo?

Several complementary methods can identify DOF5.8 interaction partners:

  • Co-immunoprecipitation (Co-IP):

    • Use DOF5.8 antibodies to pull down protein complexes

    • Identify interacting proteins by mass spectrometry

    • Validate key interactions by reciprocal Co-IP or western blotting

  • Proximity-based labeling techniques:

    • Generate DOF5.8 fusions with BioID or TurboID enzymes

    • These enzymes biotinylate proteins in close proximity to DOF5.8 in vivo

    • Identify biotinylated proteins by streptavidin pulldown and mass spectrometry

  • Split reporter systems:

    • Bimolecular Fluorescence Complementation (BiFC)

    • Split-luciferase complementation

    • Test interactions with candidate proteins such as other transcription factors or auxin signaling components

  • Yeast two-hybrid screening followed by in planta validation

When investigating interactions, focus on components of the auxin signaling pathway, as DOF5.8 modulates auxin response and affects the expression of auxin-associated transcription factors like DORNRöSCHEN and SHI-RELATED SEQUENCE 5 .

How can I distinguish between different post-translational modifications of DOF5.8?

Analyzing DOF5.8 post-translational modifications requires specialized approaches:

  • Phosphorylation analysis:

    • Generate phospho-specific antibodies against predicted phosphorylation sites

    • Use Phos-tag™ SDS-PAGE to separate phosphorylated from non-phosphorylated forms

    • Validate with phosphatase treatment controls

    • Perform mass spectrometry following immunoprecipitation to identify specific phosphorylation sites

  • Other modifications (SUMOylation, ubiquitination):

    • Use antibodies against these modifications in co-immunoprecipitation experiments

    • Perform size shift analysis under denaturing conditions

    • Verify with deubiquitinating enzyme or SUMO protease treatments

  • Functional validation:

    • Generate transgenic plants expressing site-directed mutants (e.g., S→A or S→D mutations)

    • Compare phenotypic effects and transcriptional outputs

    • Assess DNA binding capacity through EMSA or ChIP

Post-translational modifications may regulate DOF5.8 activity, stability, or interactions with other proteins, potentially explaining the complex roles of DOF5.8 in vascular development.

What cell-type specific approaches can resolve DOF5.8 expression patterns in complex tissues?

To achieve cell-type resolution in DOF5.8 studies:

  • Fluorescence-activated cell sorting (FACS) approaches:

    • Generate plants expressing fluorescent markers under cell-type specific promoters

    • Sort cells expressing DOF5.8 using FACS technology

    • Analyze protein or RNA from purified cell populations

  • Laser capture microdissection:

    • Section fixed tissue samples

    • Isolate specific cell types using laser capture technology

    • Extract proteins for western blot analysis

  • Single-cell proteomics:

    • Dissociate tissues into single cells

    • Apply mass spectrometry techniques optimized for small sample sizes

    • Quantify DOF5.8 levels in individual cells

  • High-resolution immunohistochemistry:

    • Use confocal or super-resolution microscopy

    • Combine with computational image analysis

    • Counterstain with cell-type specific markers

Cell TypeExpected DOF5.8 ExpressionDetection MethodNotes
Provascular cellsHighImmunofluorescence, FACSPrimary site of expression
Developing xylemMedium-lowIn situ immunolocalizationExpression decreases during differentiation
Mature vascular tissueLow/NoneWestern blotLimited expression in mature tissues
Meristematic regionsVariableCell sorting + Western blotContext-dependent expression

How can I resolve inconsistent DOF5.8 antibody staining patterns in immunolocalization experiments?

Inconsistent staining can result from multiple factors:

  • Tissue fixation optimization:

    • Test different fixatives (4% paraformaldehyde, paraformaldehyde/glutaraldehyde combinations)

    • Optimize fixation time (30 minutes to 4 hours depending on tissue thickness)

    • Consider vacuum infiltration to improve fixative penetration

  • Antigen retrieval methods:

    • Heat-induced epitope retrieval (10mM citrate buffer, pH 6.0)

    • Enzymatic retrieval (proteinase K treatment)

    • Detergent permeabilization optimization

  • Antibody incubation conditions:

    • Dilution series to determine optimal concentration

    • Incubation time and temperature optimization

    • Blocking buffer composition (BSA, normal serum, milk proteins)

  • Signal amplification:

    • Tyramide signal amplification for low-abundance proteins

    • Secondary antibody selection (consider using highly cross-adsorbed antibodies)

    • Fluorophore selection based on tissue autofluorescence properties

  • Controls:

    • dof5.8 mutant tissues as negative controls

    • DOF5.8 overexpression lines as positive controls

    • Pre-immune serum and secondary-only controls

What are the best approaches for quantifying DOF5.8 protein levels across different experimental conditions?

For accurate quantification:

  • Western blot quantification:

    • Use standard curves with recombinant protein

    • Include multiple biological and technical replicates

    • Apply appropriate normalization (total protein or validated reference proteins)

    • Use digital imaging systems with linear dynamic range

  • ELISA-based methods:

    • Develop sandwich ELISA with DOF5.8 antibodies

    • Include standard curves for absolute quantification

    • Optimize blocking and washing conditions to reduce background

  • Targeted mass spectrometry:

    • Develop selected reaction monitoring (SRM) or parallel reaction monitoring (PRM) assays

    • Use synthetic isotope-labeled peptides as internal standards

    • Focus on DOF5.8-specific peptides identified by discovery proteomics

  • Statistical considerations:

    • Perform power analysis to determine appropriate sample size

    • Use appropriate statistical tests for comparative analyses

    • Account for biological variability between plant specimens

How can I resolve contradictions between DOF5.8 protein levels and observed phenotypes?

When protein levels don't correlate with phenotypic effects:

  • Consider activity-level regulation:

    • Post-translational modifications may alter DOF5.8 activity without changing total protein levels

    • DNA binding capacity may be affected by chromatin accessibility

    • Interactions with cofactors may modulate function

  • Investigate temporal dynamics:

    • Perform time-course experiments to capture transient expression peaks

    • Early expression might trigger developmental cascades with delayed outcomes

    • Use inducible systems to manipulate DOF5.8 levels at specific developmental windows

  • Examine cell-type specific effects:

    • Bulk measurements might obscure cell-type specific changes

    • Use cell sorting or single-cell approaches for higher resolution

    • Consider non-cell-autonomous effects (signaling between tissues)

  • Evaluate genetic redundancy:

    • Other Dof family members may compensate for DOF5.8 function

    • Generate and analyze mutants in multiple Dof genes (DOF4.7, DOF5.1, DOF5.6)

    • Compare expression patterns and functional overlap

What bioinformatic approaches can help interpret DOF5.8 ChIP-seq data?

For comprehensive ChIP-seq data analysis:

  • Quality control and preprocessing:

    • Assess sequencing quality and depth

    • Filter low-quality reads and remove PCR duplicates

    • Align to appropriate reference genome

  • Peak calling and annotation:

    • Use algorithms like MACS2 for peak identification

    • Compare signal to appropriate controls (input DNA, IgG ChIP)

    • Annotate peaks relative to genomic features (promoters, enhancers)

  • Motif analysis:

    • Perform de novo motif discovery in peak regions

    • Compare with known Dof binding motifs

    • Analyze motif distribution and conservation

  • Integration with other datasets:

    • Correlate binding sites with gene expression data

    • Overlap with chromatin accessibility maps (ATAC-seq, DNase-seq)

    • Compare with binding profiles of related transcription factors

  • Network analysis:

    • Identify co-regulated genes

    • Perform Gene Ontology enrichment analysis

    • Construct gene regulatory networks

How can I integrate DOF5.8 protein data with transcriptomic profiles to understand its regulatory function?

For multi-omic integration:

  • Correlation analysis:

    • Compare DOF5.8 protein levels with transcript levels of potential target genes

    • Identify genes whose expression correlates with DOF5.8 abundance or activity

    • Focus on auxin-related genes and vascular development regulators

  • Perturbation experiments:

    • Analyze transcriptomes after DOF5.8 induction or repression

    • Use time-course experiments to distinguish direct from indirect targets

    • Compare with transcriptomes of dof5.8 mutants and overexpression lines

  • Network reconstruction:

    • Integrate protein-protein interaction data with transcriptional networks

    • Use algorithms like GENIE3 or WGCNA for network inference

    • Validate key regulatory relationships experimentally

  • Functional enrichment:

    • Analyze biological processes enriched among DOF5.8 targets

    • Compare with known vascular development pathways

    • Identify novel connections to other developmental processes

What standards should be applied when comparing results between different antibody-based detection methods for DOF5.8?

For reliable method comparison:

  • Antibody standardization:

    • Use the same antibody across methods when possible

    • Validate each antibody in the context of specific applications

    • Document lot-to-lot variation and include controls

  • Sample preparation consistency:

    • Use standardized protocols for tissue collection and processing

    • Process paired samples simultaneously

    • Document all deviations from standard protocols

  • Quantification approaches:

    • Use appropriate controls for each method

    • Account for differences in detection sensitivity

    • Apply method-specific normalization procedures

  • Data reporting:

    • Report all experimental details necessary for reproduction

    • Include raw data when possible

    • Clearly state limitations of each method

  • Cross-validation:

    • Confirm key findings with at least two independent methods

    • Use orthogonal techniques (antibody-based and tag-based)

    • Generate correlation plots between measurement techniques

Detection MethodSensitivitySpecificitySpatial ResolutionBest Applications
Western blottingMediumMedium-HighNoneProtein level quantification
ImmunofluorescenceMediumMedium-HighCellular/subcellularLocalization studies
ChIP-qPCRHighHighGenomic regionsTargeted binding analysis
ChIP-seqHighMedium-HighGenome-wideGlobal binding profile
Flow cytometryMedium-HighMediumCell populationQuantification in specific cell types

How can CRISPR/Cas9 genome editing enhance DOF5.8 antibody-based research?

CRISPR/Cas9 technology offers several advantages for DOF5.8 studies:

  • Endogenous tagging:

    • Insert epitope tags or fluorescent proteins at the native DOF5.8 locus

    • Maintain natural expression patterns and regulatory mechanisms

    • Allow detection with highly specific commercial antibodies against tags

  • Protein domain analysis:

    • Generate precise deletions or mutations in specific domains

    • Create allelic series to dissect protein function

    • Test requirements for protein-protein interactions or DNA binding

  • Validation resources:

    • Create knockout lines as negative controls for antibody validation

    • Generate point mutations in antibody epitopes to confirm specificity

    • Develop reporter lines for live imaging of DOF5.8 expression

  • Multi-gene manipulation:

    • Target multiple DOF family members simultaneously

    • Create higher-order mutants to address redundancy

    • Multiplex editing to study pathway components

What new approaches can resolve DOF5.8 regulatory dynamics at single-cell resolution?

Emerging technologies for single-cell analysis include:

  • Single-cell proteomics:

    • Mass cytometry (CyTOF) with metal-conjugated antibodies

    • Microfluidic antibody-based detection systems

    • Nanoscale imaging mass spectrometry

  • Spatial transcriptomics integration:

    • Combine in situ hybridization with immunodetection

    • Correlate DOF5.8 protein levels with target gene expression

    • Apply computational methods to integrate spatial datasets

  • Live cell imaging:

    • Develop split fluorescent protein systems for monitoring interactions

    • Use optogenetic tools to manipulate DOF5.8 activity with spatiotemporal precision

    • Apply FRET sensors to detect conformation changes or modifications

  • Advanced microscopy:

    • Super-resolution techniques (STORM, PALM) for precise localization

    • Light sheet microscopy for whole-tissue imaging with cellular resolution

    • Correlative light and electron microscopy for ultrastructural context

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.