HDAC8 Human

Histone Deacetylase 8 Human Recombinant
Shipped with Ice Packs
In Stock

Description

HDAC8 modulates both histone and non-histone substrates:

Histone Targets

  • Deacetylates histone H4 at lysine 382 (H4K382ac), promoting chromatin condensation and transcriptional repression .

Non-Histone Targets

SubstrateFunctional RoleDisease Association
SMC3Regulates sister chromatid cohesion during DNA repair .Cornelia de Lange Syndrome (CdLS) .
ERRα/PGC1αModulates mitochondrial biogenesis and metabolism .Metabolic disorders .
α-SMAAssociates with smooth muscle cytoskeleton .Vascular remodeling in hypertension .

Cancer

  • Oncogenic Role: HDAC8 overexpression drives proliferation in T-cell lymphoma, neuroblastoma, and acute myeloid leukemia by repressing tumor suppressors (e.g., p53) and promoting replication stress survival .

  • Therapeutic Target: Selective HDAC8 inhibitors (e.g., PCI-34051) induce apoptosis in cancer cells and synergize with checkpoint kinase inhibitors (e.g., CHK1/ATR inhibitors) to collapse replication forks .

Non-Cancer Diseases

  • CdLS: Mutations in HDAC8 disrupt SMC3 deacetylation, causing cohesinopathy and developmental defects .

  • Pulmonary Arterial Hypertension (PAH): HDAC8 upregulation in pulmonary arteries exacerbates vascular smooth muscle cell proliferation and remodeling .

HDAC8 Inhibition in Cancer Therapy

A 2024 study demonstrated that HDAC8 inhibition destabilizes replication forks by hyperacetylating SMC3, leading to R-loop accumulation and DNA damage . Combined HDAC8/CHK1 inhibition showed synthetic lethality in cancer cell lines and patient-derived xenografts :

ModelTreatmentOutcome
T-cell lymphomaHDAC8i + CHK1i85% reduction in tumor volume .
Neuroblastoma PDXHDAC8i (ACY-241)Prolonged survival (>60 days) .

Expression and Tissue Distribution

HDAC8 is predominantly expressed in smooth muscle cells, myoepithelial cells, and myofibroblasts . Immunohistochemical studies reveal cytoplasmic localization in vascular and visceral smooth muscle tissues, correlating with α-SMA expression .

Future Directions

  • Isoform-Selective Inhibitors: Developing HDAC8-specific inhibitors to minimize off-target effects .

  • Combination Therapies: Exploiting synthetic lethality with replication stress-targeting agents .

  • Biomarker Development: Correlating HDAC8 expression levels with therapeutic response in PAH and CdLS .

Product Specs

Introduction
Histone deacetylase 8 (HDAC8) is a class 1 histone deacetylase that plays a crucial role in gene regulation. It participates in skull development and regulates the ERR-alpha/PGC1-alpha transcriptional complex, which is involved in metabolism. HDAC8 modifies histones, proteins that organize DNA and control gene accessibility. This modification process, known as acetylation/deacetylation, impacts DNA structure and the binding of transcription factors, ultimately influencing gene expression.
Description
Recombinant human HDAC8, produced in Sf9 insect cells using a baculovirus expression system, is a glycosylated protein with a molecular weight of 42.6 kDa. It encompasses amino acids 1-377 of the full-length protein and includes a 6-amino acid His-tag at the C-terminus for purification. The protein is purified to a high degree using proprietary chromatographic methods.
Physical Appearance
A clear solution that has been sterilized by filtration.
Formulation
The HDAC8 protein is provided at a concentration of 0.25 mg/ml in a buffer solution containing 20mM Tris-HCl (pH 8.0), 20% glycerol, 0.1M NaCl, and 1mM DTT.
Stability
For short-term storage (up to 4 weeks), the HDAC8 solution can be kept at 4°C. For extended storage, it is recommended to freeze the solution at -20°C. To preserve protein stability during long-term storage, consider adding a carrier protein like HSA or BSA (0.1%). It's important to avoid repeated freezing and thawing of the solution.
Purity
The purity of the HDAC8 protein is greater than 80%, as determined by SDS-PAGE analysis.
Synonyms
Histone deacetylase 8, HD8, HDAC8, HDACL1, CDA07, RPD3.
Source

Sf9, Baculovirus cells.

Amino Acid Sequence
MEEPEEPADS GQSLVPVYIY SPEYVSMCDS LAKIPKRASM VHSLIEAYAL HKQMRIVKPK VASMEEMATF HTDAYLQHLQ KVSQEGDDDH PDSIEYGLGY DCPATEGIFD YAAAIGGATI TAAQCLIDGM CKVAINWSGG WHHAKKDEAS GFCYLNDAVL GILRLRRKFE RILYVDLDLH HGDGVEDAFS FTSKVMTVSL HKFSPGFFPG TGDVSDVGLG KGRYYSVNVP IQDGIQDEKY YQICESVLKE VYQAFNPKAV VLQLGADTIA GDPMCSFNMT PVGIGKCLKY ILQWQLATLI LGGGGYNLAN TARCWTYLTG VILGKTLSSE IPDHEFFTAY GPDYVLEITP SCRPDRNEPH RIQQILNYIK GNLKHVVHHH HHH.

Q&A

What is the structural organization of human HDAC8?

Human HDAC8 folds into a single α/β domain similar to bacterial HDAC-like proteins. The crystal structure reveals a zinc-binding site essential for catalytic activity, along with two potassium ions, one of which interacts with key catalytic residues . Circular dichroism data suggests potassium plays a direct role in HDAC8 fold stabilization . Significant differences from bacterial homologs are observed in the length and structure of loops surrounding the active site . The full-length protein consists of 377 amino acids with a predicted molecular mass of 45 kDa . The unique structural features of HDAC8, including its metal ion coordination sites, provide opportunities for the development of selective inhibitors.

How is HDAC8 enzymatic activity regulated in human cells?

HDAC8 activity is regulated through multiple mechanisms, with post-translational modifications playing a key role. Protein Kinase A (PKA)-mediated phosphorylation of HDAC8 at Ser39 negatively regulates its enzymatic activity . This phosphorylation leads to hyperacetylation of histones H3 and H4, though the precise biological consequences remain under investigation . Additionally, the interaction between phosphorylated HDAC8 and human EST1B (hEST1B) represents another regulatory mechanism, where HDAC8 protects hEST1B from ubiquitin-mediated degradation by the E3 ubiquitin ligase CHIP . This regulation is dependent on the phosphorylation state of HDAC8, demonstrating how post-translational modifications can alter not only catalytic activity but also protein-protein interactions.

What is the tissue distribution of HDAC8 in humans?

HDAC8 mRNA is expressed across multiple human tissues, including liver, heart, brain, lung, pancreas, placenta, prostate, and kidney . This widespread distribution suggests broad physiological roles beyond specific tissue functions. While predominantly nuclear (consistent with its histone deacetylase function), HDAC8 has also been detected in the cytoplasm of certain cell types, such as human smooth muscle cells . This dual localization pattern suggests that HDAC8 may have distinct functions depending on its subcellular compartmentalization, potentially targeting different substrate pools in the nucleus versus the cytoplasm.

What approaches can be used to establish HDAC8-expressing cell models?

To generate stable cell lines expressing human HDAC8, researchers can transfect an expression vector containing the HDAC8 coding sequence (such as p3XFlag-CMV-HDAC8) into target cells using lipid-based transfection methods . After transfection, cells should be cultured with an appropriate antibiotic selection agent (e.g., 400 μg/ml G418) for approximately 2 weeks until resistant colonies appear . Individual colonies can then be isolated, expanded, and maintained in medium with reduced antibiotic concentration (e.g., 200 μg/ml G418) . Expression of the HDAC8 protein should be verified by Western blotting and functional assays to confirm enzymatic activity. Commercial vectors with fully sequenced HDAC8 ORFs are available for researchers, with options for tagged or untagged versions depending on experimental requirements .

How does phosphorylation of HDAC8 modulate its interaction with binding partners?

The phosphorylation of HDAC8 at Ser39 by Protein Kinase A creates a molecular switch that alters its interaction profile with other proteins. Research has demonstrated that phosphorylated HDAC8 preferentially recruits Hsp70 to a complex that inhibits the CHIP-mediated ubiquitination and degradation of hEST1B . This phosphorylation-dependent interaction represents a non-catalytic function of HDAC8 in protein stabilization. To investigate such interactions experimentally, researchers have employed bacterial two-hybrid systems modified to detect phosphorylation-dependent interactions . When HDAC8 was used as bait in such a system, human EST1B was identified as a protein that specifically interacts with phosphorylated HDAC8 . This interaction was further validated using co-immunoprecipitation studies with phosphomimetic and phospho-resistant HDAC8 mutants to confirm the phosphorylation dependency.

What is the significance of HDAC8 in cancer biology and therapeutic targeting?

Knockdown of HDAC8 by RNA interference inhibits growth of human lung, colon, and cervical cancer cell lines, highlighting the importance of this HDAC subtype for tumor cell proliferation . This finding positions HDAC8 as a potential therapeutic target in multiple cancer types. The availability of the crystal structure of human HDAC8 in complex with hydroxamic acid inhibitors provides a foundation for structure-based drug design of selective HDAC8 inhibitors . The unique features of HDAC8, including its potassium binding sites and distinct active site loop structures, offer opportunities for developing inhibitors with improved selectivity over other HDAC family members. Such selective inhibitors could potentially reduce side effects associated with pan-HDAC inhibition while maintaining efficacy against HDAC8-dependent tumors.

How do metal ions contribute to HDAC8 structure and function?

HDAC8 is a zinc-dependent enzyme with a catalytic zinc ion essential for its deacetylase activity . Additionally, the crystal structure of human HDAC8 reveals two potassium ions that are not present in bacterial HDAC-like proteins . One of these potassium ions interacts directly with key catalytic residues, suggesting a role in the catalytic mechanism . Circular dichroism data indicates that potassium ions play a direct role in stabilizing the tertiary structure of HDAC8 . These findings highlight the importance of considering metal ion coordination when designing experiments to study HDAC8 activity or developing inhibitors. For recombinant protein production, appropriate metal supplementation in expression and purification buffers is critical to ensure properly folded and catalytically active enzyme.

What computational approaches can identify novel HDAC8 substrates?

Structure-based computational methods have been developed to predict potential HDAC8 substrates from the human proteome. The Rosetta FlexPepBind protocol evaluates peptide-binding ability to the HDAC8 active site based on structural models of this interaction . This approach uses peptide sequences extracted from proteins with known acetylated sites to identify those that fit well into the HDAC8 binding pocket . The computational predictions can be validated through in vitro deacetylation assays using synthetic acetylated peptides and recombinant HDAC8 . This integrated computational-experimental approach has identified many new in vitro HDAC8 peptide substrates, expanding our understanding of HDAC8's substrate specificity beyond histones . Such approaches are valuable for systematically mapping the full spectrum of HDAC8 substrates in the human acetylome.

How does HDAC8 influence telomere maintenance through EST1B?

HDAC8 interacts with human EST1B (hEST1B), a telomerase-associated factor, in a phosphorylation-dependent manner . This interaction protects hEST1B from ubiquitin-mediated degradation by the E3 ubiquitin ligase CHIP . Phosphorylated HDAC8 preferentially recruits Hsp70 to a complex that inhibits CHIP-mediated ubiquitination of hEST1B . Given that hEST1B is a telomerase-associated factor, this finding suggests a potential role for HDAC8 in telomere maintenance. The regulation of hEST1B stability by HDAC8 provides a mechanistic link between HDAC8 function and telomere biology, which could have implications for cellular senescence, aging, and cancer. This represents an important non-catalytic function of HDAC8 that extends its biological significance beyond histone deacetylation.

What expression systems are optimal for producing recombinant human HDAC8?

For bacterial expression of human HDAC8, cDNAs encoding the full-length protein can be amplified by PCR and inserted into appropriate expression vectors such as p3XFlag-CMV . When expressing HDAC8 in mammalian cells, vectors containing strong promoters like CMV with selectable markers (e.g., neomycin resistance) are commonly used . For stable expression, transfected cells should be selected with appropriate antibiotics (G418 at 400 μg/ml initially, reduced to 200 μg/ml for maintenance) . Special consideration should be given to metal supplementation in growth media and purification buffers, as HDAC8 requires zinc for catalytic activity and potassium for structural stability . For studying phosphorylation-dependent functions, co-expression with appropriate kinases (e.g., PKA) or the use of phosphomimetic mutations (S39D) can be employed.

What approaches can identify direct HDAC8 substrates versus indirect effects?

To identify direct HDAC8 substrates, a multi-faceted approach combining computational prediction and experimental validation is recommended. Computational methods like Rosetta FlexPepBind can provide initial predictions based on structural compatibility with the HDAC8 active site . In vitro deacetylation assays using recombinant HDAC8 and synthetic acetylated peptides can validate these predictions biochemically . For cellular validation, acetylome profiling using mass spectrometry to compare acetylation patterns in HDAC8-depleted versus control cells can identify differentially acetylated proteins. To distinguish direct from indirect effects, catalytically inactive HDAC8 mutants (H142A/H143A) can be used as negative controls. Additionally, time-course experiments following acute HDAC8 inhibition can help identify immediate (likely direct) versus delayed (potentially indirect) acetylation changes.

What techniques are suitable for studying HDAC8-mediated protein-protein interactions?

To identify proteins that interact with HDAC8, both unbiased and targeted approaches can be employed. Modified bacterial two-hybrid systems have been used successfully to screen for proteins that interact with HDAC8 in a phosphorylation-dependent manner . For validating specific interactions, co-immunoprecipitation experiments using epitope-tagged HDAC8 (e.g., Flag-HDAC8) can be performed . When studying phosphorylation-dependent interactions, phosphomimetic (S39D) and phospho-resistant (S39A) HDAC8 mutants provide valuable experimental tools . For mapping interaction domains, truncation mutants of both HDAC8 and its binding partners can identify the minimal regions required for interaction. Proximity-based labeling techniques such as BioID or APEX fused to HDAC8 can identify proteins that are in close proximity to HDAC8 in living cells, potentially revealing novel interaction partners.

MethodApplicationAdvantagesLimitations
Bacterial Two-HybridInitial screeningCan detect phospho-dependent interactionsMay miss transient interactions
Co-ImmunoprecipitationValidation of specific interactionsPreserves native complexesMay lose weak interactions during washing
BioID/APEXIn situ proximity mappingIdentifies interactions in living cellsLabels proximity not just direct interactions
Yeast Two-HybridLarge-scale screeningHigh throughputHigh false positive/negative rates
Protein ComplementationLive cell interaction studiesReal-time monitoringMay affect protein folding

How can researchers evaluate HDAC8 catalytic activity in vitro and in cells?

For in vitro assessment of HDAC8 enzymatic activity, fluorogenic substrate assays using commercially available kits provide a straightforward approach. Alternatively, synthetic acetylated peptides derived from known or putative substrates can be incubated with recombinant HDAC8, followed by mass spectrometry analysis to quantify deacetylation . When evaluating HDAC8 activity in cells, Western blotting with acetyl-specific antibodies against known HDAC8 substrates can track changes in acetylation status. For more comprehensive analysis, global acetylome profiling using mass spectrometry following HDAC8 inhibition or depletion can identify affected substrates. Activity-based probes that covalently bind to active HDAC8 can be used to assess the active fraction of HDAC8 in complex biological samples. When comparing wild-type and mutant HDAC8 variants, circular dichroism spectroscopy should be employed to ensure proper protein folding, as activity differences could stem from structural perturbations rather than specific residue functions.

What considerations are important when designing HDAC8 inhibitor studies?

When conducting HDAC8 inhibitor studies, several key considerations should be addressed. Selectivity assessment should include testing against a panel of other HDAC family members to determine the specificity profile. Structure-activity relationship studies should leverage the unique features of HDAC8, including the potassium binding sites and distinct active site loop structures . For cellular studies, target engagement should be confirmed using cellular thermal shift assays (CETSA) or monitoring the acetylation status of known HDAC8 substrates. Dose-response relationships should be established, and appropriate treatment durations determined based on the pharmacokinetic properties of the inhibitor and the biological process being studied. Negative controls should include structurally related compounds that lack HDAC8 inhibitory activity. For validating inhibitor-induced phenotypes, genetic approaches (HDAC8 knockdown/knockout) should produce similar effects if the inhibitor is specific.

How might the unique features of HDAC8 be exploited for developing isoform-selective inhibitors?

The crystal structure of human HDAC8 reveals several unique features that distinguish it from other HDAC family members, including distinct active site loop structures and potassium binding sites . These structural differences can be exploited for developing selective HDAC8 inhibitors. Structure-based drug design approaches can target specific residues or pockets unique to HDAC8. The potassium binding sites, particularly the one that interacts with catalytic residues, might be targeted to develop allosteric inhibitors that disrupt the metal coordination network . Additionally, the foot pocket region of HDAC8, which differs from other HDACs, offers another site for selective targeting. Fragment-based drug discovery approaches can identify novel chemical scaffolds with inherent selectivity for HDAC8. The development of such selective inhibitors would enable more precise targeting of HDAC8-dependent processes while minimizing off-target effects on other HDACs, potentially leading to improved therapeutic windows for clinical applications.

What is the potential significance of non-canonical HDAC8 functions beyond histone deacetylation?

Emerging evidence indicates that HDAC8 possesses important functions beyond its canonical role in histone deacetylation. The interaction between phosphorylated HDAC8 and hEST1B reveals a role in protecting this telomerase-associated factor from ubiquitin-mediated degradation . This function appears to be independent of HDAC8's catalytic activity and instead relies on its ability to form specific protein complexes in a phosphorylation-dependent manner. Additionally, the growing list of non-histone HDAC8 substrates suggests roles in multiple cellular processes beyond transcriptional regulation . Future research should investigate whether HDAC8 possesses other non-canonical functions, such as scaffolding roles in signaling complexes or regulation of protein stability through mechanisms independent of deacetylase activity. Understanding these non-canonical functions could reveal new therapeutic opportunities and explain phenotypes that cannot be attributed solely to changes in histone acetylation.

How do genetic variations in HDAC8 contribute to human disease phenotypes?

Several synonyms for HDAC8 in clinical contexts include CDLS5 (Cornelia de Lange Syndrome 5) and MRXS6 (Mental Retardation, X-linked, Syndromic 6), suggesting genetic associations with specific disorders . Future research should comprehensively analyze how mutations or polymorphisms in HDAC8 correlate with disease phenotypes. Structure-function studies can determine how specific mutations affect HDAC8 catalytic activity, protein-protein interactions, or subcellular localization. Patient-derived cells carrying HDAC8 mutations can be studied to identify dysregulated pathways and potential therapeutic targets. CRISPR-based approaches to introduce specific HDAC8 mutations into model organisms or cell lines can establish causality between genetic variations and phenotypic outcomes. Additionally, exploring whether HDAC8 variants affect responsiveness to HDAC inhibitors could have implications for personalized medicine approaches in conditions where HDAC8 dysfunction plays a role.

What role does HDAC8 play in the crosstalk between different epigenetic regulatory mechanisms?

HDAC8's function in histone deacetylation positions it as a key player in the broader landscape of epigenetic regulation. Future research should investigate how HDAC8 activity coordinates with other epigenetic mechanisms such as DNA methylation, histone methylation, and chromatin remodeling. ChIP-seq studies for HDAC8 combined with other epigenetic marks could reveal genomic regions where multiple epigenetic mechanisms converge. Proteomics approaches could identify interactions between HDAC8 and other epigenetic regulators, potentially revealing novel complexes. The impact of HDAC8 inhibition or depletion on global epigenetic landscapes, including changes in other histone modifications or DNA methylation patterns, would provide insights into regulatory hierarchies. Understanding these crosstalks could reveal synthetic lethal interactions between HDAC8 inhibition and modulation of other epigenetic pathways, potentially leading to new combination therapeutic strategies in diseases with epigenetic dysregulation.

How might HDAC8 contribute to cellular stress responses and environmental adaptations?

HDAC8 is involved in stress response according to the search results , but the specific mechanisms remain to be fully elucidated. Future research should investigate how HDAC8 activity, localization, or interactions change under various stress conditions (oxidative stress, DNA damage, hypoxia, nutrient deprivation). Time-course studies following stress induction could reveal whether HDAC8 is an early or late responder in stress pathways. The role of post-translational modifications, particularly phosphorylation at Ser39, should be examined in the context of stress signaling. Identification of stress-specific HDAC8 substrates could reveal how HDAC8 contributes to cellular adaptation. Additionally, exploring whether HDAC8 inhibition sensitizes cells to specific stressors could have therapeutic implications, particularly in cancer where stress response pathways are often dysregulated. Understanding HDAC8's role in cellular resilience could open new avenues for targeting stress adaptation mechanisms in disease settings.

Product Science Overview

Introduction

Histone deacetylase 8 (HDAC8) is a member of the class I histone deacetylases (HDACs), which are crucial enzymes involved in the transcriptional regulation of gene expression in eukaryotic cells . HDACs catalyze the removal of acetyl groups from lysine residues on histones, leading to chromatin condensation and transcriptional repression .

Structure and Function

HDAC8 is a 43 kDa protein that is expressed in various tissues and has been shown to play a role in several biological processes . It functions on both histone and non-histone substrates, producing free lysine and acetate by catalyzing the hydrolysis of acetyllysine side chains . HDAC8 is involved in skull morphogenesis and the metabolic control of the ERR-alpha/PGC1-alpha transcriptional complex .

Pathological Role

HDAC8 has been implicated in various diseases, particularly cancer. Overexpression and deregulation of HDAC8 contribute to cancer cell proliferation, metastasis, immune evasion, and drug resistance . Additionally, HDAC8 is associated with non-cancer diseases such as Cornelia de Lange Syndrome (CdLS), infectious diseases, cardiovascular diseases, pulmonary diseases, and myopathy .

Therapeutic Potential

Given its involvement in multiple diseases, HDAC8 is considered an attractive therapeutic target. Various HDAC8 selective inhibitors (HDAC8is) have been developed, showing promising anti-cancer effects . These inhibitors aim to restore normal acetylation levels, thereby reversing the pathological phenotypes associated with HDAC8 deregulation .

Recombinant HDAC8

Recombinant human HDAC8 is produced using baculovirus-infected insect cells and is available in carrier-free formulations . This recombinant protein is used in various research applications, including studying the enzyme’s activity and screening for potential inhibitors .

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.