Histone deacetylase 8 (HDAC8) is a class I histone deacetylase encoded by the Hdac8 gene located on the X chromosome in mice (MGI:1917565) . It regulates chromatin structure and gene expression by removing acetyl groups from lysine residues on histones and non-histone substrates. Mouse models have been instrumental in elucidating HDAC8’s roles in development, reproduction, cancer, and genome stability. Key findings from knockout (KO) and conditional KO models reveal its tissue-specific functions and potential therapeutic implications.
HDAC8 is critical for cranial neural crest cell (NCC) patterning and skull morphogenesis. Global Hdac8 deletion in mice causes perinatal lethality due to skull instability, brain hemorrhage, and soft tissue herniation . Conditional deletion in NCCs phenocopies this defect, demonstrating HDAC8’s specific role in repressing homeobox transcription factors (e.g., Otx2, Lhx1) that disrupt frontal skull formation .
HDAC8 ensures proper spindle assembly and chromosome alignment during oocyte maturation. Depletion or inhibition of HDAC8 in mouse oocytes results in:
Spindle defects: 56.45% of HDAC8-depleted oocytes show abnormal spindle morphology vs. 21.04% in controls .
Aneuploidy: 51.65% of MII eggs from HDAC8-depleted oocytes are aneuploid vs. 23.76% in controls .
Stage-specific deletions reveal that HDAC8 is essential early in oogenesis. Vasa-Cre-mediated KO (pre-meiotic S phase) causes subfertility due to smaller oocytes with reduced meiotic competence, whereas Zp3-Cre (post-meiotic) KO preserves fertility .
HDAC8 drives tumor progression and therapy resistance in mouse models:
Neuroblastoma: Selective HDAC8 inhibition (e.g., PCI-34051) reduces tumor growth by 60% in xenografts, increases differentiation markers (neurofilament), and induces caspase-3-mediated apoptosis .
Breast cancer metastasis: HDAC8 suppresses SIRT7 via SMAD3/4 complexes, hyperactivating TGF-β signaling to promote lung metastasis .
Replication stress: Combined HDAC8 and checkpoint kinase inhibition (e.g., AZD-7762) causes replication fork collapse, synergistically killing cancer cells in patient-derived xenografts (PDXs) .
HDAC8 inhibition reprograms immune-excluded tumors into immunogenic niches. In hepatocellular carcinoma (HCC) mouse models:
Histone deacetylase 8 (HDAC8) is a class 1 member of the histone deacetylase/acuc/apha family. This enzyme plays a crucial role in skull development and the regulation of the ERR-alpha/PGC1-alpha transcriptional complex, which is involved in metabolic processes. Histones, proteins that interact with DNA, are essential for controlling gene expression, cell cycle progression, and developmental processes. The acetylation and deacetylation of histones modify the structure of chromosomes, influencing how easily transcription factors can access and regulate DNA.
Recombinant HDAC8 Mouse, produced in Sf9 Baculovirus cells, is a single, glycosylated polypeptide chain. It comprises 383 amino acids (with amino acids 1-377 being specifically identified) and has a molecular weight of 42.5 kDa. Under reducing conditions on SDS-PAGE, it migrates between 40-57 kDa. This HDAC8 protein is expressed with a 6 amino acid His tag at the C-terminus and purified using proprietary chromatographic techniques.
The HDAC8 protein solution is supplied at a concentration of 0.25 mg/ml. It is prepared in a buffer consisting of Phosphate Buffered Saline at a pH of 7.4 and contains 10% glycerol.
Analysis by SDS-PAGE indicates that the purity of this product is greater than 90%.
Histone deacetylase 8, HD8, Hdac8.
MEMPEEPANS GHSLPPVYIY SPEYVSICDS LVKVPKRASM VHSLIEAYAL HKQMRIVKPK VASMEEMATF HTDAYLQHLQ KVSQEGDEDH PDSIEYGLGY DCPATEGIFD YAAAIGGGTI TAAQCLIDGK CKVAINWSGG WHHAKKDEAS GFCYLNDAVL GILRLRRKFD RILYVDLDLH HGDGVEDAFS FTSKVMTVSL HKFSPGFFPG TGDMSDVGLG KGRYYSVNVP IQDGIQDEKY YHICESVLKE VYQAFNPKAV VLQLGADTIA GDPMCSFNMT PVGIGKCLKY VLQWQLATLI LGGGGYNLAN TARCWTYLTG VILGKTLSSE IPDHEFFTAY GPDYVLEITP SCRPDRNEPH RIQQILNYIK GNLKHVVHHH HHH.
HDAC8 functions as a lysine deacetylase, specifically responsible for deacetylating the cohesin subunit SMC3. This enzymatic activity is critical for recycling cohesin from one cell cycle to the next . Unlike other class I HDACs that have been extensively studied, HDAC8's specific role in physiological processes like adult neurogenesis has only recently been investigated . The enzyme requires a single zinc ion for the hydrolysis of N6-acetyllysine side chains in acetylated protein substrates such as SMC3, with critical amino acid residues (D178, H180, and D267) coordinating the zinc ion and stabilizing the active site structure .
HDAC8 exhibits a distinctive expression pattern in the subventricular zone (SVZ) of the adult mouse brain. Immunofluorescence studies have demonstrated that HDAC8 is highly expressed in Nestin-positive neural stem/progenitor cells . Further characterization using cell-type specific markers revealed HDAC8 expression across various neural progenitor subtypes, including GFAP-positive radial glia-like cells, MASH1-positive transient amplifying cells, and doublecortin (Dcx)-positive neuroblast cells . This widespread expression across the neural stem cell lineage suggests HDAC8 plays important regulatory roles throughout the process of adult neurogenesis.
Conditional knockout of HDAC8 specifically in Nestin-positive neural stem/progenitor cells leads to significant phenotypic effects in adult mice. The most notable consequences include:
Inhibition of neural stem/progenitor cell proliferation in the SVZ
Reduced neural differentiation capacity
Decreased expression of neural stem/progenitor cell markers such as GFAP and Nestin
These phenotypic changes were confirmed through immunofluorescence staining of adult brain sections using antibodies against neural stem/progenitor cell markers . The specificity of the knockout was validated by the loss of HDAC8 immunoreactivity in the SVZ of Nestin-CreERT2; Hdac8 flox/flox mice following tamoxifen administration .
When designing conditional HDAC8 knockout experiments, researchers should consider the following methodological approach:
Genetic Strategy: Utilize the Cre-loxP system with Hdac8 flox/flox mice crossed with an appropriate Cre driver line. For neural stem cell studies, Nestin-CreERT2 mice have proven effective for tamoxifen-inducible deletion of Hdac8 specifically in Nestin-positive cells .
Induction Protocol: Administer tamoxifen according to a carefully planned timeline. As demonstrated in previous studies, tamoxifen administration in adult Nestin-CreERT2; Hdac8 flox/flox mice allows for temporal control of gene deletion .
Validation Methods: Confirm successful deletion through immunostaining for HDAC8 in target tissues, comparing knockout regions with control areas within the same tissue section .
Phenotypic Analysis: Perform comprehensive phenotypic assessment including immunofluorescence staining with cell-type-specific markers (GFAP, Nestin, DCX) to evaluate effects on different neural cell populations .
Functional Testing: Include behavioral assays relevant to the neurological functions potentially affected by HDAC8 deletion.
When designing HDAC8 inhibitor studies in mouse models, researchers should consider:
Inhibitor Selection: Choose inhibitors with demonstrated specificity for HDAC8. PCI-34051 has been validated as a selective HDAC8 inhibitor in neural stem cell studies , while compound 4b has shown efficacy in Huntington's disease models through preferential targeting of HDAC1 and HDAC3 .
Dose-Response Assessment: Test multiple concentrations to establish dose-dependent effects. For instance, studies with HDAC inhibitor 4b in N171-82Q transgenic mice examined various dosages to determine optimal therapeutic effects while minimizing potential toxicity .
Treatment Duration: Consider both acute and chronic administration protocols. In Huntington's disease studies, long-term administration was necessary to observe significant improvements in body weight, motor function, and cognitive performance .
Delivery Method: Select appropriate administration routes (intraperitoneal injection, oral gavage, etc.) based on the pharmacokinetic properties of the inhibitor and the target tissue accessibility.
Control Groups: Include both vehicle-treated and untreated groups to control for effects of the delivery vehicle itself.
To accurately assess HDAC8 enzymatic activity in mouse tissues, researchers should implement the following methodological approaches:
Enzyme Purification: Express wild-type and mutated forms of HDAC8 (for comparison studies) in a suitable expression system such as E. coli, followed by protein purification .
Activity Assays: Utilize established commercial HDAC8 assays that measure deacetylase activity through fluorescent or colorimetric substrates. Previous studies have demonstrated the reliability of these assays for measuring relative activities of wild-type versus mutant HDAC8 proteins .
Tissue-Specific Analysis: For in vivo studies, prepare tissue homogenates under conditions that preserve enzymatic activity, controlling for potential confounding factors such as endogenous inhibitors.
Structure-Function Correlation: When investigating specific mutations, map these onto the known crystal structure of HDAC8 to predict functional consequences, as done with various HDAC8 mutations in previous studies .
Substrate Specificity Assessment: Consider using physiologically relevant substrates such as acetylated SMC3 peptides to better approximate in vivo activity.
When analyzing transcriptional data from HDAC8 inhibition or knockout experiments, researchers should:
Pathway Analysis: Apply robust bioinformatics approaches to identify affected gene networks. Previous studies revealed that HDAC8 inhibition in adult SVZ cells impacts cytokine-mediated signaling and cell cycle pathways . Similarly, HDAC8 inhibitor 4b treatment in Huntington's disease mouse models affected post-translational modification pathways, including protein phosphorylation and ubiquitination .
Validation Techniques: Confirm key differentially expressed genes using real-time qPCR. Studies have validated changes in genes such as Ube2K, Ubqln, Ube2e3, Usp28, and Sumo2 following HDAC inhibitor treatment .
Temporal Analysis: Consider time-dependent changes in gene expression to distinguish primary from secondary effects of HDAC8 inhibition.
Cell-Type Specificity: When working with heterogeneous tissues like brain, consider the contribution of different cell populations to the observed transcriptional changes.
Integration with Functional Data: Correlate transcriptional changes with observed phenotypic alterations to establish causality. For example, downregulation of cell cycle pathways following HDAC8 inhibition correlates with reduced neurosphere size in adult mouse SVZ cultures .
When comparing HDAC8 inhibition versus genetic knockout approaches, researchers should consider:
Completeness of Inhibition: Pharmacological inhibition rarely achieves 100% blockade of enzymatic activity, whereas genetic knockout can completely eliminate protein expression. This difference may account for quantitative differences in observed phenotypes.
Temporal Dynamics: Genetic knockouts (particularly constitutive ones) affect development from early stages, while inhibitor treatments can be initiated at specific timepoints. Inducible systems like the Nestin-CreERT2 allow for temporal control of genetic deletion, approximating the timing flexibility of inhibitor administration .
Compensatory Mechanisms: Long-term genetic deletion may trigger compensatory upregulation of related HDACs, which might not occur with acute pharmacological inhibition.
Off-Target Effects: Inhibitors may affect other proteins despite their selectivity, complicating interpretation. Careful comparison between inhibitor studies and genetic models can help identify potential off-target effects.
Dose-Dependent Effects: Inhibitors allow for dose-response studies, which can reveal threshold effects not easily observed in genetic models. For example, different doses of HDACi 4b showed varying efficacy in ameliorating Huntington's disease phenotypes .
The correlation between mouse HDAC8 deficiency and human HDAC8 mutations reveals important translational insights:
Neurodevelopmental Effects: HDAC8 mutations in humans cause a phenotypic spectrum including Cornelia de Lange syndrome (CdLS)-like features, ocular hypertelorism, and large fontanelle . In mice, HDAC8 deficiency affects neural stem cell proliferation and differentiation , suggesting conserved roles in neurodevelopment across species.
Sex-Specific Effects: Human HDAC8 mutations show X-linked inheritance patterns with more severe phenotypes in males than females . This sex-weighted effect is evidenced by differences in average height, weight, and head circumference between males and females with HDAC8 mutations .
X-Inactivation Patterns: In female patients with HDAC8 mutations, severely skewed X-inactivation (>95:5) is observed in peripheral blood, suggesting strong selection against cells expressing the mutant allele . This should be considered when designing and interpreting mouse models with heterozygous females.
Enzymatic Activity Correlation: The severity of clinical phenotypes in humans correlates with the degree of loss of HDAC8 enzymatic activity . Mutations that completely abolish activity (such as H71Y, H180R, and G304R) affect residues that are identical from humans through yeast and generally result in more severe phenotypes .
For investigating HDAC8 function in neural stem cells, researchers should consider these methodological approaches:
Neurosphere Assays: Neurosphere cultures from the adult mouse SVZ provide an effective model system for studying HDAC8 function in neural stem/progenitor cells. Treatment with selective HDAC8 inhibitors such as PCI-34051 has been shown to reduce neurosphere size, reflecting decreased proliferation capacity .
Primary Neural Stem Cell Cultures: Isolated primary NSCs/NPCs maintained under defined conditions can be utilized to study cell-autonomous effects of HDAC8 manipulation through genetic approaches (siRNA, CRISPR) or pharmacological inhibition.
Differentiation Assays: Sequential differentiation protocols that recapitulate the transition from neural stem cells to neurons provide a platform to assess HDAC8's role in neuronal differentiation and maturation.
Live Cell Imaging: Time-lapse microscopy of labeled neural stem cells following HDAC8 manipulation can reveal dynamic effects on proliferation kinetics, cell division modes, and differentiation trajectories.
Co-culture Systems: Co-culturing HDAC8-manipulated neural stem cells with other cell types (astrocytes, microglia) can help dissect cell-autonomous versus non-cell-autonomous effects of HDAC8 dysfunction.
When designing HDAC8 inhibitor studies, researchers should implement the following controls:
Vehicle Controls: Include matched vehicle-treated groups that receive the same solvent/carrier as the inhibitor treatment group to control for potential effects of the delivery vehicle itself.
Dosage Controls: Test multiple dosages to establish dose-response relationships and determine minimal effective concentrations. Studies with HDACi 4b tested different doses to establish optimal therapeutic effects in Huntington's disease models .
Pan-HDAC Inhibitor Comparison: Include a pan-HDAC inhibitor treatment group to distinguish HDAC8-specific effects from general HDAC inhibition effects.
Genetic Validation: When possible, compare pharmacological inhibition results with genetic models (HDAC8 knockout or knockdown) to confirm target specificity.
Biochemical Validation: Measure HDAC8 activity in target tissues to confirm successful inhibition at the biochemical level, correlating enzyme inhibition with observed phenotypes.
Time Course Controls: Sample at multiple timepoints to distinguish immediate from delayed effects and to monitor potential development of tolerance or compensatory mechanisms.
For robust assessment of HDAC8 expression in mouse brain tissues, researchers should consider:
Immunohistochemistry/Immunofluorescence: Combined with cell-type-specific markers (Nestin, GFAP, MASH1, Dcx) to characterize expression across different neural cell populations . This approach has successfully demonstrated HDAC8 expression in various neural progenitor subtypes in the SVZ.
Western Blotting: For quantitative comparison of HDAC8 protein levels across different brain regions or experimental conditions, using appropriate loading controls and validated antibodies.
RT-qPCR: To measure HDAC8 mRNA expression levels, particularly useful for detecting changes in transcriptional regulation of the HDAC8 gene.
Single-Cell RNA Sequencing: Provides high-resolution cell-type-specific expression data, allowing identification of HDAC8 expression patterns across the full spectrum of neural cell types.
In Situ Hybridization: For spatial localization of HDAC8 mRNA in intact brain tissue sections, complementing protein-level studies.
ChIP-Seq Analysis: To identify genomic regions directly regulated by HDAC8, providing insights into the mechanism of action in specific cell types.
Region-specific conditional knockout approaches offer several advantages for advancing HDAC8 research:
Circuit-Specific Effects: By targeting HDAC8 deletion to specific brain circuits (hippocampus, cortex, striatum), researchers can dissect region-specific functions in learning, memory, motor control, and other behaviors.
Developmental Timing: Combining region-specific Cre drivers with inducible systems allows manipulation of HDAC8 at different developmental stages, revealing potential developmental windows of vulnerability.
Adult Neurogenesis Focus: Targeted deletion in neurogenic niches (SVZ, subgranular zone) can further elucidate HDAC8's role in adult neurogenesis without confounding effects from other brain regions .
Disease Modeling: Region-specific deletion relevant to particular neurological disorders (e.g., striatum for Huntington's disease models) can enhance translational relevance of findings.
Compensation Assessment: Comparing phenotypes between global versus region-specific knockouts can reveal compensatory mechanisms that may occur in a region-specific manner.
Current mouse model research suggests several promising therapeutic applications for HDAC8 inhibitors:
Neurodevelopmental Disorders: Given HDAC8's role in human conditions like Cornelia de Lange syndrome , targeted inhibition might modulate developmental trajectories in relevant mouse models of neurodevelopmental disorders.
Neurodegenerative Diseases: HDAC inhibitors have shown beneficial effects in Huntington's disease mouse models , potentially through modulation of the ubiquitin-proteasomal and autophagy pathways that affect accumulation and clearance of disease-related proteins.
Brain Injury Recovery: Considering HDAC8's involvement in neural stem cell regulation , inhibitors might promote neural regeneration following traumatic brain injury or stroke.
Cancer Therapeutics: Increased HDAC8 expression is associated with poor outcomes in neuroblastoma, and HDAC8 inhibition reduces cell proliferation in neuroblastoma models , suggesting potential applications in brain cancer treatment.
Cognitive Enhancement: By modulating epigenetic regulation in adult neural stem cells, HDAC8 inhibitors might enhance cognitive function in conditions associated with cognitive decline.
To advance understanding of HDAC8 substrate specificity in the physiological context, researchers should consider:
Proteomics Approaches: Employ mass spectrometry-based proteomics to identify acetylated proteins in wild-type versus HDAC8-deficient tissues, focusing on changes in acetylation status of potential substrates.
HDAC8 Interactome Analysis: Use co-immunoprecipitation followed by mass spectrometry to identify proteins that physically interact with HDAC8 in different cell types, potentially revealing context-specific substrates.
Activity-Based Protein Profiling: Develop activity-based probes for HDAC8 that can label active enzyme in live cells and tissues, allowing visualization and isolation of the enzyme in its native context.
Substrate-Trapping Mutants: Generate catalytically inactive HDAC8 mutants that can bind but not process substrates, effectively "trapping" them for identification.
In Vivo Acetylome Analysis: Compare the acetylome of specific cell populations (e.g., neural stem cells) with and without HDAC8 activity, using cell type-specific isolation techniques combined with acetyl-proteomics.
Substrate Validation: Confirm identified substrates through targeted approaches such as site-directed mutagenesis of acetylation sites and functional studies to determine the biological significance of HDAC8-mediated deacetylation.
Histone deacetylase 8 (HDAC8) is an enzyme that plays a crucial role in the regulation of gene expression through the modification of chromatin structure. This enzyme is part of the histone deacetylase family, which is involved in the removal of acetyl groups from lysine residues on histone proteins. The mouse recombinant version of HDAC8 is a valuable tool for research, providing insights into its function and potential therapeutic applications.
HDAC8 belongs to class I of the histone deacetylase family. The recombinant mouse HDAC8 is produced in Sf9 Baculovirus cells and consists of a single, glycosylated polypeptide chain containing 383 amino acids, with a molecular mass of approximately 42.5 kDa . This enzyme is known for its histone deacetylase activity, which involves the removal of acetyl groups from histone proteins, leading to a more condensed chromatin structure and repression of gene transcription .
Histone deacetylation by HDAC8 plays a critical role in various cellular processes, including transcriptional regulation, cell cycle progression, and developmental events . By altering the acetylation status of histones, HDAC8 affects the accessibility of transcription factors to DNA, thereby influencing gene expression. This enzyme is also involved in skull morphogenesis and the metabolic control of the ERR-alpha/PGC1-alpha transcriptional complex .
HDAC8 has been implicated in several disease states, particularly in cancer. Overexpression and deregulation of HDAC8 are associated with cancer cell proliferation, metastasis, immune evasion, and drug resistance . Additionally, HDAC8 is linked to the pathogenesis of neuroblastoma, acute myeloid leukemia, and other cancers . Beyond cancer, HDAC8 is also involved in non-cancer diseases such as Cornelia de Lange Syndrome (CdLS), cardiovascular diseases, pulmonary diseases, and myopathy .
Given its significant role in various diseases, HDAC8 is considered an attractive therapeutic target. Researchers have developed several HDAC8 selective inhibitors (HDAC8is) that have shown promising anti-cancer effects . These inhibitors work by specifically targeting HDAC8, thereby reversing the pathological phenotypes associated with its overexpression and deregulation.