HDAC7 Antibody Pair

Shipped with Ice Packs
In Stock

Product Specs

Buffer
**Capture Buffer:** 50% Glycerol, 0.01M PBS, pH 7.4
**Detection Buffer:** 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
We typically dispatch orders within 1-3 business days of receipt. Delivery times may vary depending on the shipping method and destination. For specific delivery timeframes, please contact your local distributor.
Notes
For optimal results, we recommend using the capture antibody at a concentration of 0.5 µg/mL and the detection antibody at a concentration of 0.34 µg/mL. It is essential to determine the optimal dilutions experimentally based on your specific research needs.
Synonyms
Histone deacetylase 7A,HDAC7,HDAC7A
Target Names
HDAC7

Q&A

What is HDAC7 and why is it important in research?

HDAC7 (Histone Deacetylase 7) is a class IIa histone deacetylase responsible for the removal of acetyl groups from lysine residues on histones and non-histone proteins. It functions primarily as a signal-dependent repressor of gene transcription.

HDAC7 has emerged as a critical regulatory protein in multiple biological processes:

  • T cell development and selection processes

  • Neuronal survival mechanisms

  • Cancer progression in multiple tumor types

  • Inflammatory responses in macrophages

Methodologically, studying HDAC7 requires specific antibodies that recognize distinct domains or isoforms. Current research indicates HDAC7 has multiple isoforms resulting from alternative splicing, with the inclusion or exclusion of exon 9 (HDAC7-E9) being particularly important for T cell function .

How do HDAC7 antibody pairs differ from single antibodies for detection?

An HDAC7 antibody pair consists of two antibodies that bind to different epitopes on the HDAC7 protein:

  • Capture antibody: Typically immobilized on a solid support to bind and isolate HDAC7 from complex samples. For example, the commercially available rabbit polyclonal antibody directed against human HDAC7 .

  • Detection antibody: Typically labeled (directly or indirectly) for visualization or quantification, such as the mouse monoclonal anti-HDAC7 IgG2b kappa .

The methodological advantage of using antibody pairs includes:

  • Increased specificity through dual epitope recognition

  • Enhanced sensitivity for quantification in complex biological samples

  • Reduced background noise in assays like ELISA

  • Ability to detect conformational changes by targeting different domains

When designing experiments, researchers should select antibody pairs that:

  • Recognize distinct, non-overlapping epitopes

  • Don't compete for antigen binding

  • Maintain reactivity under assay conditions (e.g., pH, salt concentration)

  • Detect the specific isoform of interest

What are the key domains of HDAC7 that should be considered when selecting antibodies?

HDAC7 contains several functional domains that should be considered when selecting antibodies:

DomainFunctionConsideration for Antibody Selection
N-terminal domain (aa 1-513)Regulatory region; contains phosphorylation sites and 14-3-3 binding motifsCritical for detecting regulation by phosphorylation and association with 14-3-3 proteins
Catalytic domain (C-terminal)Deacetylase activityImportant for studying enzymatic function; some HDAC7 functions are catalytically independent
Exon 9 regionAlternative splicing affects protein stability and interaction with 14-3-3 proteinsEssential for distinguishing between isoforms (HDAC7iE9 vs. HDAC7ΔE9)

Methodological approach:

  • For studying phosphorylation-dependent nuclear export: Select antibodies recognizing the N-terminal domain

  • For differentiation between isoforms: Use antibodies specific to regions affected by alternative splicing

  • For enzymatic activity studies: Choose antibodies that don't interfere with the catalytic domain

  • Consider using domain-specific antibodies such as those targeting HDAC7(1-519) or HDAC7(514-953)

How can HDAC7 antibody pairs be optimized for studying protein-protein interactions in T cell signaling?

HDAC7 interactions with 14-3-3 proteins are critical for its function in T cell signaling. Research has shown that alternative splicing of HDAC7 exon 9 regulates this interaction .

Methodological approach for studying these interactions:

  • Co-immunoprecipitation (Co-IP) optimization:

    • Use antibodies targeting the N-terminal region of HDAC7 where 14-3-3 binding occurs

    • Pre-clear lysates thoroughly to reduce non-specific binding

    • Include phosphatase inhibitors to preserve phosphorylation-dependent interactions

    • Consider mild detergents (0.1% NP-40) to maintain native protein complexes

  • Proximity ligation assay (PLA) protocol:

    • Fix cells with 4% paraformaldehyde (10 min, RT)

    • Permeabilize with 0.2% Triton X-100 (10 min, RT)

    • Block with 5% BSA (1 hour, RT)

    • Incubate with primary antibodies against HDAC7 and 14-3-3 (overnight, 4°C)

    • Follow manufacturer's protocol for PLA probes and detection

  • Bimolecular Fluorescence Complementation (BiFC):

    • Tag HDAC7 and potential interaction partners with complementary fluorescent protein fragments

    • Express in relevant T cell models

    • Monitor reconstitution of fluorescence upon protein interaction

Research data indicates that HDAC7iE9 has approximately double the association with 14-3-3 proteins compared to HDAC7ΔE9 under stimulated conditions , making this interaction a critical target for investigation.

What methods are most effective for studying HDAC7 in cancer progression models?

HDAC7 has been implicated in multiple cancers, including choroidal melanoma , nasopharyngeal carcinoma , and non-small cell lung cancer (NSCLC) . Effective study methods include:

  • Expression analysis in tumor tissues:

    • Immunohistochemistry (IHC) protocols:

      • FFPE section preparation: 4-6 μm thickness

      • Antigen retrieval: Citrate buffer (pH 6.0), 95°C, 15 min

      • Primary antibody incubation: Anti-HDAC7 (1:100-1:200), overnight at 4°C

      • Detection: HRP-conjugated secondary and DAB substrate

  • Functional studies in cancer models:

    • shRNA-mediated knockdown using validated sequences:

      • shHdac7-1 (TRCN0000039335)

      • shHdac7-2 (TRCN0000039337)

    • Overexpression models using:

      • Full-length HDAC7

      • Domain-specific constructs (N-terminal or C-terminal)

      • Phosphorylation site mutants

  • Downstream target analysis:

    • ChIP-seq protocol optimization:

      • Crosslinking: 1% formaldehyde, 10 min, RT

      • Sonication: Optimize to achieve 200-500 bp fragments

      • IP: Anti-HDAC7 antibody (4-5 μg per reaction)

      • Controls: IgG control and input samples

Research has shown that HDAC7 promotes cancer progression through multiple mechanisms:

  • In choroidal melanoma: HDAC7/c-Myc signaling pathway promotes proliferation

  • In nasopharyngeal carcinoma: HDAC7 downregulates miR-4465 and subsequently upregulates EphA2

  • In NSCLC: HDAC7 interacts with β-catenin, causing decreased acetylation at Lys49 and decreased phosphorylation at Ser45

How can researchers effectively distinguish between HDAC7 isoforms resulting from alternative splicing?

Alternative splicing of HDAC7, particularly of exon 9, significantly impacts protein function. Effective methods to distinguish between isoforms include:

  • RT-PCR based detection:

    • Design primers flanking alternatively spliced regions

    • For HDAC7 exon 9: Use low-cycle RT-PCR

    • Quantify inclusion as percent spliced in (PSI) values

    • RT-PCR can detect changes as small as 21% in exon inclusion

  • Isoform-specific antibody selection:

    • Choose antibodies raised against splice junction sequences

    • Validate specificity using recombinant isoform proteins

    • Consider creating custom antibodies if commercial options are unavailable

  • Protein stability analysis:

    • Treat cells with cycloheximide (CHX) to block translation

    • Monitor protein degradation over time by western blot

    • The HDAC7iE9 isoform shows approximately double the stability of HDAC7ΔE9 under stimulated conditions

IsoformExon 9 Status14-3-3 BindingProtein StabilityLEF1 Binding
HDAC7iE9IncludedHigher~2x longer half-lifeLower
HDAC7ΔE9ExcludedLowerShorter half-lifeHigher

Research has demonstrated a bimodal pattern of HDAC7 exon 9 splicing regulation during T cell stimulation, with initial reduction in the first 12 hours followed by steady increase to approximately 50% inclusion by 48 hours .

What validation steps should be performed to confirm HDAC7 antibody specificity?

Thorough validation of HDAC7 antibodies is critical for reliable research outcomes:

  • Western blot validation:

    • Positive controls: Cell lines with known HDAC7 expression (K-562 cells)

    • Negative controls: HDAC7 knockout cells generated by CRISPR-Cas9

    • Size verification: Expected molecular weight is approximately 102-103 kDa

    • Multiple antibody comparison: Test different clones targeting distinct epitopes

  • Immunoprecipitation validation:

    • Pull-down efficiency assessment

    • Mass spectrometry confirmation of precipitated protein

    • Reciprocal IP with interacting partners (e.g., 14-3-3 proteins)

    • Competition assays with recombinant protein

  • Immunofluorescence validation:

    • Subcellular localization verification

    • Comparison with GFP-tagged HDAC7 expression

    • Signal abolishment after HDAC7 knockdown or knockout

    • Peptide competition assays

  • Cross-reactivity assessment:

    • Testing against other HDAC family members (particularly class IIa HDACs)

    • Species cross-reactivity testing if working with multiple model organisms

    • Signal detection in tissues known to express or lack HDAC7

Proper antibody validation should also include verification across different experimental conditions, such as stimulated versus unstimulated T cells, where HDAC7 expression and localization may change .

What are the optimal conditions for using HDAC7 antibody pairs in co-immunoprecipitation studies?

Co-immunoprecipitation (Co-IP) is a powerful technique for studying HDAC7 interactions. Optimal conditions include:

  • Lysis buffer optimization:

    • For HDAC7-14-3-3 interactions: Use mild conditions

      • 50 mM Tris-HCl (pH 7.4)

      • 150 mM NaCl

      • 1% NP-40 or 0.5% Triton X-100

      • Protease and phosphatase inhibitor cocktails

    • For chromatin-associated interactions: Consider including nuclease treatment

  • Antibody selection and immobilization:

    • Use FLAG-tagged HDAC7 constructs for clean pull-downs

    • For endogenous HDAC7: 0.5-4.0 μg antibody per 1.0-3.0 mg of total protein lysate

    • Pre-clear lysates with protein A/G beads (1 hour, 4°C)

    • Covalently cross-link antibodies to beads to prevent antibody contamination in eluates

  • Washing and elution conditions:

    • Perform 4-5 washes with decreasing salt concentrations

    • Consider stringent washes only for final wash

    • Elute with either low pH, competitive peptides, or SDS sample buffer

  • Controls and validation:

    • Input control (typically 5-10% of lysate used for IP)

    • IgG control (same species as IP antibody)

    • Reciprocal IP where possible

    • Consider sequential IP for complex interactions

Research using these approaches has successfully demonstrated increased association of 14-3-3 proteins with HDAC7iE9 versus HDAC7ΔE9, while HDAC7ΔE9 associates more efficiently with LEF1 .

How can researchers accurately measure changes in HDAC7 protein levels and activity in response to stimuli?

Accurately measuring HDAC7 changes requires combining protein level and activity assessments:

  • Protein level quantification:

    • Western blot analysis:

      • Loading controls: β-actin, GAPDH (cytoplasmic), Lamin B1 (nuclear)

      • Quantification: Densitometry with normalization to loading controls

      • Subcellular fractionation: Separate nuclear and cytoplasmic fractions to track shuttling

    • Flow cytometry for single-cell analysis:

      • Fix cells with 4% PFA (10 min, RT)

      • Permeabilize with 0.1% saponin or 0.1% Triton X-100

      • Block with 2% BSA (30 min, RT)

      • Incubate with anti-HDAC7 (1:100, 1 hour, RT)

      • Wash and incubate with fluorophore-conjugated secondary antibody

  • HDAC7 activity assessment:

    • Commercial HDAC activity assays with HDAC7 immunoprecipitates

    • Substrate specificity considerations:

      • HDAC7 shows preference for specific acetylated substrates

      • Some HDAC7 functions are deacetylase-independent

  • Phosphorylation status monitoring:

    • Phospho-specific antibodies or Phos-tag gels

    • Mass spectrometry analysis of phosphorylation sites

    • Treatment with phosphatase inhibitors during cell lysis

  • Time-course experiments:

    • HDAC7 shows dynamic regulation:

      • Exon 9 inclusion follows a bimodal pattern during T cell stimulation

      • Protein levels increase 2.7-fold following stimulation

      • Major protein increase occurs between 12-24 hours post-stimulation

Research has shown that HDAC7 levels and activity must be monitored over appropriate time courses, as the full biological response may take 24-48 hours to develop .

What are common pitfalls when studying HDAC7 in primary immune cells?

Primary immune cells present specific challenges for HDAC7 research:

  • Cell isolation and viability issues:

    • Problem: Low cell yields and viability affect protein detection

    • Solution: Optimize isolation protocols with minimal mechanical stress; include DNase I treatment for sticky chromatin; use viability dyes to gate viable cells

  • Rapid changes in HDAC7 expression and localization:

    • Problem: HDAC7 undergoes dynamic regulation during immune cell activation

    • Solution: Implement precise time-course experiments; observe bimodal pattern of exon 9 splicing (initial decrease followed by increase)

  • Isoform-specific detection challenges:

    • Problem: Antibodies may not distinguish between splice variants

    • Solution: Use RT-PCR to correlate with protein detection; generate isoform-specific antibodies; use tagged constructs for validation

  • Stimulus-dependent variability:

    • Problem: Different stimuli may cause varying HDAC7 responses

    • Solution: Standardize activation protocols; test multiple activation pathways (e.g., PMA vs. TCR engagement); include appropriate time points (12h, 24h, 48h)

  • Nuclear-cytoplasmic shuttling complications:

    • Problem: HDAC7 localization changes affect extraction efficiency

    • Solution: Use subcellular fractionation protocols; include phosphatase inhibitors to preserve phosphorylation states; validate with immunofluorescence

Research has shown that during T cell stimulation, HDAC7 undergoes complex regulatory changes including transcriptional upregulation (~3.2-fold), altered splicing patterns, and increased protein stability .

How can researchers design experiments to study the deacetylase-independent functions of HDAC7?

HDAC7 exhibits important deacetylase-independent functions that require specific experimental approaches:

  • Catalytic mutant expression systems:

    • Generate HDAC7 constructs with mutations in catalytic residues

    • Express wild-type vs. catalytically dead mutants

    • Compare phenotypic outcomes and interactome differences

    • Research shows HDAC7-mediated neuroprotection doesn't require its catalytic domain

  • Domain-specific construct utilization:

    • Express N-terminal domain HDAC7(1-519) or C-terminal domain HDAC7(514-953)

    • Analyze domain-specific protein interactions

    • Compare transcriptional effects through RNA-seq or targeted gene expression analysis

  • Experimental design considerations:

    • Include HDAC inhibitor controls (TSA, MS-275, SAHA)

    • Use isoform-specific constructs (HDAC7iE9 vs. HDAC7ΔE9)

    • Compare effects in the presence/absence of specific signaling pathway inhibitors

    • Validate with HDAC7 knockdown/knockout systems

  • Readout selection:

    • For neuroprotective functions: Measure apoptosis and c-jun expression

    • For T cell development: Monitor surface markers (CD3, CD28, CD69)

    • For cancer studies: Assess proliferation and metastasis markers

Research has demonstrated that HDAC7-mediated neuroprotection occurs via the inhibition of c-jun expression, acting at the transcriptional level through direct association with the c-jun promoter .

What methodological approaches are recommended for studying HDAC7 in the context of cancer therapeutics?

HDAC7's role in multiple cancers makes it a potential therapeutic target. Recommended methodological approaches include:

  • Inhibitor screening and evaluation:

    • HDAC7-selective inhibition assessment:

      • In vitro enzymatic assays with recombinant HDAC7

      • Cellular target engagement assays

      • Correlation with phenotypic outcomes

    • Combination therapy evaluation:

      • Study synergy with established cancer therapeutics

      • Determine combination indexes using Chou-Talalay method

      • Evaluate effects on both cancer and normal cells

  • Patient-derived models:

    • Primary patient sample testing:

      • Compare HDAC7 expression in tumor vs. normal tissues

      • Correlate expression with clinical parameters

      • Test ex vivo drug sensitivity

    • Patient-derived xenografts (PDX):

      • Establish PDX models from different cancer types

      • Validate HDAC7 expression and function

      • Test targeted therapies

  • Biomarker development:

    • Identify response predictors:

      • Correlation of HDAC7 expression with patient outcomes

      • Isoform-specific expression analysis

      • Identification of HDAC7-dependent gene signatures

  • Resistance mechanism investigation:

    • Study adaptive responses:

      • HDAC7 expression changes following treatment

      • Alternative splicing modifications

      • Compensatory pathway activation

Research has shown significant correlation between HDAC7 expression and poor prognosis in multiple cancer types, including choroidal melanoma , nasopharyngeal carcinoma , and NSCLC . For example, in NSCLC, elevated expression of HDAC7 was positively correlated with poor prognosis, TNM stage, and tumor differentiation .

What controls are essential when using CRISPR-edited cell lines for HDAC7 research?

CRISPR-edited cell lines require rigorous controls for valid HDAC7 research:

  • Edit verification controls:

    • Genomic verification:

      • PCR and sequencing of targeted region

      • Off-target analysis by whole-genome sequencing

    • Transcript verification:

      • RT-PCR across edited region

      • RNA-seq for splice variant analysis

    • Protein verification:

      • Western blot for complete protein loss (knockout)

      • Domain-specific antibodies for truncation verification

  • Functional compensation controls:

    • Expression analysis of other HDAC family members:

      • Class IIa HDACs (HDAC4, 5, 9) may compensate for HDAC7 loss

      • qRT-PCR and protein analysis of related HDACs

    • Rescue experiments:

      • Wild-type HDAC7 re-expression

      • Isoform-specific rescue (HDAC7iE9 vs. HDAC7ΔE9)

      • Domain-specific constructs

  • Phenotypic validation:

    • Cell line-specific phenotype verification:

      • Proliferation assays

      • Migration/invasion assessments

      • Target gene expression analysis

    • Multiple clone analysis to control for clonal effects

    • Compare to shRNA knockdown phenotypes using validated sequences:

      • shHdac7-1 (TRCN0000039335)

      • shHdac7-2 (TRCN0000039337)

  • Temporal considerations:

    • Acute vs. chronic HDAC7 loss comparison

    • Inducible systems for temporal control

    • Analysis of adaptive responses over time

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 2024 Thebiotek. All Rights Reserved.