Cyclin H forms a ternary complex with cyclin-dependent kinase 7 (CDK7) and MAT1, functioning as a CDK-activating kinase (CAK). This complex phosphorylates CDK2 and CDC2 to regulate cell cycle progression . Additionally, it is a component of the TFIIH transcription factor, enabling RNA polymerase II-mediated transcription initiation . Key interactions include:
CCNH Human is utilized in:
Kinase activation assays: Measures CAK activity using substrates like CDK2 .
Antibody development: Mouse monoclonal antibodies (e.g., clone PAT3G6AT) are generated for Western blot and ELISA .
NanoBRET® target engagement: Co-expressed with kinase-NanoLuc fusions to study intracellular kinase-ligand binding .
Cancer relevance: Cyclin H overexpression correlates with tumor progression due to dysregulated CDK activation .
Structural insights: The His-tagged recombinant protein retains native conformation, confirmed by SDS-PAGE and circular dichroism .
Therapeutic targeting: Inhibitors of the Cyclin H-CDK7 complex show promise in preclinical cancer models .
MGSSHHHHHH SSGLVPRGSH MYHNSSQKRH WTFSSEEQLA RLRADANRKF RCKAVANGKV LPNDPVFLEP HEEMTLCKYY EKRLLEFCSV FKPAMPRSVV GTACMYFKRF YLNNSVMEYH PRIIMLTCAF LACKVDEFNV SSPQFVGNLR ESPLGQEKAL EQILEYELLL IQQLNFHLIV HNPYRPFEGF LIDLKTRYPI LENPEILRKT ADDFLNRIAL TDAYLLYTPS QIALTAILSS ASRAGITMES YLSESLMLKE NRTCLSQLLD IMKSMRNLVK KYEPPRSEEV AVLKQKLERC HSAELALNVI TKKRKGYEDD DYVSKKSKHE EEEWTDDDLV ESL.
CCNH belongs to the highly conserved cyclin family and functions as a regulator of CDK kinases. It forms a complex with CDK7 kinase and ring finger protein MAT1, creating a CDK-activating kinase (CAK) that phosphorylates CDK2 and CDC2 kinases . This complex is crucial for cell cycle progression. Additionally, CCNH and its kinase partner are components of the TFIIH and RNA polymerase II protein complexes, indicating its dual role in transcriptional regulation and cell cycle control . This established connection between basal transcription processes and cell cycle machinery makes CCNH a critical factor in understanding fundamental cellular mechanisms.
CCNH expression varies significantly across human tissues, with particular elevation observed in proliferative tissues. Research methodologies for assessing this variation typically involve quantitative reverse transcription-polymerase chain reaction (qRT-PCR) to detect expression levels across different tissue samples . When investigating tissue-specific expression patterns, researchers should employ multi-tissue panels with appropriate controls and normalization methods. Comparative analysis across both normal and pathological tissues reveals that CCNH expression is often dysregulated in cancerous tissues compared to their normal counterparts, especially in lung tissues where elevated expression correlates with disease progression .
CCNH has been demonstrated to interact with several key proteins including P53, Cyclin-dependent kinase 7, and MNAT1 . These interactions form functional complexes that regulate both transcription and cell cycle progression. To investigate these protein interactions, researchers typically employ co-immunoprecipitation assays, yeast two-hybrid screening, or proximity ligation assays. The interaction between CCNH and P53 is particularly significant given P53's role as a tumor suppressor, suggesting that disruptions in this interaction may contribute to carcinogenesis. These interactions place CCNH at a critical intersection of cellular regulation pathways, making it an important target for understanding both normal cellular function and disease mechanisms.
Post-translational modifications of CCNH significantly alter its regulatory capacity within the CDK-activating kinase complex. Phosphorylation patterns particularly influence CCNH's ability to activate downstream kinases. To investigate these modifications, researchers should employ mass spectrometry approaches to identify modification sites, followed by site-directed mutagenesis to create phosphomimetic or phospho-deficient mutants. Functional assays comparing wild-type and mutant CCNH activity in reconstituted CAK complexes in vitro can reveal how specific modifications impact kinase activity. Additionally, comparing modification patterns between normal and cancer cells may identify cancer-specific regulation mechanisms that could be therapeutically targeted. These studies require careful experimental design with appropriate controls to distinguish direct effects of CCNH modifications from secondary consequences.
Distinguishing CCNH's functions in transcription versus cell cycle control requires sophisticated experimental design. A recommended approach combines CRISPR-Cas9 genome editing to create domain-specific mutations that selectively disrupt one function while preserving others. This should be coupled with ChIP-seq to map CCNH genomic binding sites and identify transcriptional targets, alongside high-resolution cell cycle analysis using flow cytometry with markers for specific cell cycle phases. Time-resolved proteomics can track CCNH complex formation throughout the cell cycle, while conditional knockdown systems allow temporal control of CCNH depletion to determine phase-specific requirements. When interpreting results, researchers should be careful to distinguish direct from compensatory effects, as disruption of one function may indirectly affect the other due to the interconnected nature of these cellular processes .
Single-cell technologies provide unprecedented insights into CCNH functional heterogeneity across cell populations. Single-cell RNA-seq and proteomics reveal cell-specific expression patterns that bulk analysis methods miss, while single-cell ATAC-seq can identify accessibility changes in CCNH-regulated genes. To implement these approaches, researchers should begin with optimized tissue dissociation protocols that preserve cellular integrity, followed by careful quality control to exclude damaged cells. Computational analysis requires specialized algorithms to handle dropout events and technical noise. Integration of multiple single-cell modalities (transcriptomics, proteomics, and epigenomics) provides the most comprehensive view of CCNH function. When interpreting results, researchers should validate key findings using spatial transcriptomics or imaging mass cytometry to preserve tissue context information that might be lost during dissociation.
Identifying CCNH-regulated genes across cancer types requires integrated multi-omics approaches. Begin with differential expression analysis comparing tissues or cells with high versus low CCNH expression, using RNA-seq data from resources like TCGA and CPTAC databases . Follow this with ChIP-seq analysis to identify direct CCNH binding sites, distinguishing direct from indirect regulatory effects. Motif enrichment analysis can identify co-regulatory factors that function alongside CCNH. When analyzing these datasets, employ appropriate statistical corrections for multiple hypothesis testing and batch effects. Pathway enrichment analysis will identify biological processes enriched among CCNH targets. To validate computational predictions, researchers should perform targeted experiments with CCNH knockdown or overexpression followed by RNA-seq or proteomics. Cross-cancer analysis reveals both shared and cancer-specific CCNH regulatory networks that may inform therapeutic targeting strategies.
Analysis of CCNH genetic variation requires specialized statistical approaches tailored to the research question. For association studies, logistic regression with covariates for population stratification is essential, while rare variant analysis demands burden tests or sequence kernel association tests that aggregate variants within functional domains. When conducting these analyses, researchers should implement rigorous quality control to filter low-quality variants and address linkage disequilibrium patterns. Functional annotation of variants using tools like PolyPhen or SIFT helps prioritize potentially damaging changes. Bayesian approaches can integrate prior biological knowledge about CCNH function to improve statistical power. For evaluating potential gene-environment interactions, researchers should employ cross-validation to avoid overfitting and use appropriate multiple testing corrections. These methodological considerations are crucial for robust interpretation of genetic association results involving CCNH across diverse population cohorts.
CCNH expression demonstrates significant correlations with treatment responses across multiple cancer therapies. Research indicates that elevated CCNH expression is associated with poor prognosis in lung cancer patients, suggesting potential treatment resistance mechanisms . To investigate these correlations, researchers should employ prospective cohort designs with pre-treatment biopsies for baseline CCNH assessment, followed by standardized response evaluation criteria. Multivariate analyses must control for confounding factors including cancer stage, grade, and concurrent mutations. Additionally, researchers should explore whether CCNH mediates response through its cell cycle regulatory function or through transcriptional regulation of resistance genes. In vitro drug sensitivity assays using isogenic cell lines with modified CCNH expression can validate clinical correlations and elucidate mechanistic relationships. These findings have important implications for personalized therapy selection and development of combination approaches targeting CCNH-related pathways.
Developing CCNH as a clinical biomarker requires rigorous methodological validation across multiple dimensions. Begin with analytical validation to establish assay precision, accuracy, and reproducibility across different laboratories. Clinical validation requires large, prospectively collected cohorts with comprehensive follow-up data to determine sensitivity, specificity, and predictive value. As demonstrated in lung cancer studies, CCNH expression correlates with poor prognosis, suggesting potential as a prognostic biomarker . Researchers must standardize specimen collection, processing, and storage protocols to minimize pre-analytical variables. Immunohistochemistry protocols require optimization of antibody specificity, dilution, and scoring systems. For circulating biomarker applications, establish appropriate detection limits and reference ranges. Additionally, researchers should determine whether CCNH alone provides sufficient clinical utility or requires integration into multi-marker panels. Economic analysis of the cost-effectiveness of CCNH testing compared to existing biomarkers is essential for clinical implementation.
BESH studies investigating CCNH function must carefully balance basic research objectives with clinical considerations. These studies meet both the definition of basic research and the NIH definition of a clinical trial . Design should focus on understanding fundamental aspects of CCNH biology without specific applications toward processes or products in mind. When designing such studies, researchers should clearly define the experimental manipulation intended to perturb CCNH-related physiological processes. Protocol development requires careful consideration of inclusion/exclusion criteria to create homogeneous study populations while maintaining generalizability. Outcome measures should focus on mechanistic biomarkers rather than clinical endpoints. Researchers must navigate the regulatory requirements, including registration and results reporting obligations that apply to all studies meeting the clinical trial definition . Additionally, statistical power calculations should account for expected variability in human samples, and analytical plans should include strategies for handling missing data and protocol deviations. These methodological considerations ensure rigorous investigation of CCNH biology in human subjects while maintaining regulatory compliance.
Establishing appropriate experimental controls is critical when modulating CCNH expression in human cell lines. For knockdown experiments using siRNA or shRNA, researchers should include both non-targeting controls and rescue experiments with siRNA-resistant CCNH constructs to confirm specificity. When using CRISPR-Cas9 genome editing, include multiple guide RNAs targeting different regions of CCNH alongside appropriate non-targeting guides. Additionally, researchers should generate clonal knockout lines with confirmed absence of CCNH protein and validate these using both sequencing and western blot. For overexpression studies, employ inducible systems to control expression levels and timing, with empty vector controls processed identically. Cell cycle synchronization protocols ensure comparable cell populations when studying cycle-dependent effects. The experimental design should include monitoring of potential compensatory mechanisms, such as upregulation of related cyclins, that might confound interpretation of results. These methodological controls are essential for generating reliable and reproducible data on CCNH function in human cell lines.
Multi-omics integration provides comprehensive insights into CCNH regulatory networks beyond what single data types can reveal. Begin with parallel genomic, transcriptomic, proteomic, and epigenomic profiling of the same samples to minimize technical variation. For computational integration, researchers can employ matrix factorization methods, network-based approaches, or Bayesian integration frameworks that account for the different statistical properties of each data type. When analyzing integrated datasets, identify convergent evidence across multiple platforms to prioritize high-confidence findings. Temporal sampling designs capture dynamic changes in CCNH networks during processes like cell cycle progression or differentiation. Visualization tools such as multi-layer network representations help communicate complex relationships across data types. Functional validation of key predictions using targeted experiments is essential to confirm computational findings. This integrated approach has successfully identified novel CCNH-regulated pathways in cancer contexts that were not apparent from single-platform studies .
Characterizing CCNH structural variations requires complementary computational and experimental approaches. Begin with in silico structural modeling using available crystal structures and homology modeling for regions lacking experimental structures. Molecular dynamics simulations can predict how variants affect protein flexibility and interaction surfaces. For experimental validation, researchers should employ circular dichroism spectroscopy to assess secondary structure changes and thermal shift assays to evaluate stability alterations. Functional consequences can be assessed through reconstituted kinase assays comparing wild-type and variant CCNH proteins. Surface plasmon resonance or isothermal titration calorimetry quantifies changes in binding affinities with known interaction partners. Cell-based assays with structure-guided mutations help connect structural changes to cellular phenotypes. When interpreting results, researchers should consider that different structural changes may selectively impact specific CCNH functions while preserving others, potentially explaining the diverse phenotypes observed in different cellular contexts following CCNH alteration .
Cyclin-H forms a trimeric complex with CDK7 and MAT1, known as the CDK-activating kinase (CAK) complex . This complex is essential for the activation of CDKs, which are critical for cell cycle progression. Cyclin-H is also a component of the transcription factor TFIIH, which is involved in transcription initiation and DNA repair .
Recombinant human Cyclin-H is typically produced using various expression systems, such as E. coli or insect cells. The recombinant protein is often tagged with a His-tag or GST-tag to facilitate purification . For example, one method involves expressing full-length human Cyclin-H in insect cells using an N-terminal GST tag . Another method involves expressing Cyclin-H with a His-tag in E. coli .