Recombinant CCND1 proteins are synthesized using heterologous expression systems:
E. coli: High-yield production with bacterial vectors (e.g., pET-28a) for structural studies, though post-translational modifications (e.g., phosphorylation) are absent .
Mammalian Cells: Systems like HEK293 or CHO cells produce glycosylated and phosphorylated CCND1, enabling functional studies of RB interaction and cell cycle regulation .
Purification involves affinity chromatography (e.g., His-tag) and size-exclusion chromatography to achieve >90% purity .
Forms complexes with CDK4/6 to phosphorylate RB, releasing E2F transcription factors and driving G1/S transition .
CCND1a enhances DNA damage response (DDR) by recruiting Rad51 to repair sites, conferring chemoresistance .
Interacts with transcriptional coactivators (e.g., STAT3, PPARγ) to regulate gene expression .
Exhibits corepressor activity on promoters of differentiation genes (e.g., INS, NEUROD1) .
Amplification: Observed in >30% of breast and mantle cell lymphomas, leading to overexpression .
Mutations: G870A mutation generates CCND1b, which paradoxically inhibits proliferation in certain contexts .
CDK4/6 inhibitors (e.g., palbociclib) disrupt CCND1-CDK4 complexes, inducing G1 arrest .
Strategies targeting CCND1 splicing (e.g., antisense oligonucleotides) aim to restore normal isoform ratios .
Used in cell cycle synchronization assays (e.g., serum deprivation followed by CCND1 addition) .
Assessed for DDR modulation in models of colon and breast cancer .
Mechanism | CCND1a | CCND1b |
---|---|---|
CDK4/6 Activation | High (drives G1/S transition) | Low (deficient in RB phosphorylation) |
DNA Damage Response | Enhances (Rad51 recruitment) | Impaired (fails to recruit Rad51) |
Apoptosis Induction | No | Yes (in cervical cancer models) |
Cancer | Primary Alteration | Clinical Impact |
---|---|---|
Mantle Cell Lymphoma | t(11;14) translocation | Overexpression drives proliferation |
Breast Cancer | Amplification | Predicts chemoresistance |
Glioblastoma | Overexpression | Correlates with poor prognosis |
Our Recombinant Human CCND1 protein is a valuable tool for cell cycle research, providing insights into crucial aspects of cell cycle progression and cellular proliferation. As a G1/S-specific cyclin-D1, encoded by the CCND1 gene, it serves as a regulatory subunit of cyclin-dependent kinases, playing a pivotal role in driving the cell cycle from G1 phase to S phase. The protein's involvement in various cancers makes it a key target in cancer research.
Produced in an E.coli expression system, our Recombinant Human CCND1 protein offers high-quality, full-length protein spanning the 1-295aa region. An N-terminal 6xHis-tag facilitates efficient protein purification, ensuring reliable and consistent results for your experiments. With a purity exceeding 90%, as determined by SDS-PAGE, our Recombinant Human CCND1 protein provides the accuracy and dependability required for your research. Choose between liquid or lyophilized powder formats to meet your specific laboratory needs and advance your cell cycle investigations.
CCND1 (also known as BCL1 or PRAD1) is encoded by a gene located on chromosome 11 and consists of 5 exons and 4 introns . The protein contains functionally distinct domains that mediate its various activities:
N-terminal domain: Mediates interactions with key proteins including RAD51, as demonstrated by experimental evidence using purified recombinant proteins. This interaction is critical for CCND1's role in DNA repair processes .
CDK-binding domain: Essential for forming active complexes with CDK4 and CDK6, which subsequently phosphorylate the retinoblastoma (Rb) protein.
C-terminal domain: Contains regulatory sequences including phosphorylation sites that control protein stability and degradation.
Methodologically, researchers can study domain-specific functions through mutational analysis, creating truncated versions of CCND1 lacking specific domains to assess their impact on various cellular processes including cell cycle progression, protein interactions, and DNA repair functions.
CCND1 produces two main isoforms: full-length CCND1a and the alternatively spliced CCND1b, which has a divergent C-terminal domain . Their differences include:
Feature | CCND1a | CCND1b |
---|---|---|
Structure | Contains complete C-terminus | Has divergent C-terminus due to alternative splicing |
Cell cycle regulation | Accelerates cell proliferation by promoting cell cycle progression | May inhibit cell cycle progression in some contexts |
DNA damage response | Confers resistance to DNA damage therapy by inducing DNA damage response | No reported role in DNA damage response |
Invasion/Metastasis | Limited role in invasion/metastasis | Promotes invasion and metastasis in breast cancer independently of CCND1a |
The G870A polymorphism is the most common mutation in the CCND1 gene and significantly affects its splicing patterns . This single nucleotide polymorphism occurs at the exon 4-intron 4 boundary and influences the production of CCND1 isoforms:
Mechanistically, the G870A mutation affects the splice donor site, altering the efficiency of normal splicing processes .
The A allele promotes alternative splicing that leads to increased production of the CCND1b isoform.
Based on analysis from the Catalogue of Somatic Mutations in Cancer and dbSNP databases, G870A is present in approximately 6% of cancer patients .
To study this polymorphism experimentally, researchers employ:
PCR-RFLP (restriction fragment length polymorphism) analysis to genotype patient samples
Minigene splicing assays to evaluate splicing efficiency in vitro
RT-PCR with isoform-specific primers to quantify the relative abundance of each isoform
The G870A mutation has been associated with increased risk in multiple cancer types including breast cancer, liver cancer, colorectal cancer, bladder cancer, endometrial cancer, and esophageal and gastric cardiac cancer .
CCND1 plays an essential role in promoting G1 to S phase transition through several coordinated mechanisms :
Assembly with CDK partners: CCND1 is synthesized during the G1 phase in response to growth factor stimulation and assembles with either CDK4 or CDK6 to form active kinase complexes .
Rb inactivation: Once formed, CCND1-CDK4/6 complexes phosphorylate the retinoblastoma (Rb) protein, inactivating its growth-suppressive function. This phosphorylation releases E2F transcription factors that activate genes required for DNA replication and S phase entry .
CDK inhibitor sequestration: CCND1-CDK complexes titrate CDK inhibitors such as p21Cip1 and p27Kip1, preventing them from inhibiting other cyclin-CDK complexes necessary for cell cycle progression .
To experimentally investigate this coordination, researchers can use:
Kinase assays with purified components to measure CCND1-CDK4/6 activity toward Rb substrates
Co-immunoprecipitation to detect complex formation between CCND1 and CDK4/6
Western blotting to monitor Rb phosphorylation status
Flow cytometry with propidium iodide staining to assess cell cycle distribution
CCND1 protein stability is tightly regulated through multiple mechanisms that directly impact cell cycle progression :
Growth factor-dependent synthesis and stability: In the absence of growth factor stimulation, CCND1 becomes unstable and undergoes ubiquitin-mediated degradation .
Phosphorylation-triggered degradation: CCND1 degradation is triggered by its phosphorylation, leading to G1/S phase arrest when growth factors are withdrawn .
Radiation-induced degradation: Research shows that radiation results in decreased total levels of CCND1, representing an important regulatory mechanism in response to DNA damage .
Researchers studying CCND1 stability can employ these methodological approaches:
Cycloheximide chase assays to measure protein half-life
Ubiquitination assays to detect polyubiquitinated CCND1
Proteasome inhibitors (e.g., MG132) to determine the contribution of proteasomal degradation
Site-directed mutagenesis of key phosphorylation sites to analyze their impact on protein stability
CCND1 dysregulation in cancer occurs through several distinct mechanisms :
Gene amplification: The most common mechanism involves amplification of the CCND1 gene (located on chromosome 11q13). Approximately 15-20% of breast cancers exhibit CCND1 amplification .
Protein overexpression: CCND1 overexpression is observed in more than 50% of breast cancers, indicating that mechanisms beyond gene amplification contribute to elevated protein levels . Overexpression can occur through enhanced transcription, increased mRNA stability, or altered protein degradation.
G870A mutation: This common polymorphism affects splicing and is associated with increased risk across multiple cancer types, including breast, liver, colorectal, bladder, and endometrial cancers .
Other mutations: Additional CCND1 mutations have been cataloged, including synonymous mutations, missense mutations, truncating mutations, and splice mutations, though these are less common than amplification or the G870A polymorphism .
For experimental detection of these alterations, researchers should employ:
FISH (Fluorescence In Situ Hybridization) for detecting gene amplification
qRT-PCR for measuring mRNA expression levels
Immunohistochemistry and Western blotting for protein expression analysis
PCR-RFLP or sequencing for detecting specific mutations like G870A
The prognostic significance of CCND1 alterations shows considerable variation across different cancer types :
Cancer Type | CCND1 Amplification | CCND1 Overexpression |
---|---|---|
ER+ Breast Cancer | Poor prognosis | Controversial (both good and poor prognosis reported) |
Head and Neck Squamous Cell Carcinoma | Poor prognosis | Poor prognosis (amplification is a more reliable marker) |
Gastric Cancer | Poor prognosis | Poor prognosis |
Esophageal Cancer | Poor prognosis | Poor prognosis |
Colorectal Cancer | Poor prognosis | Poor prognosis |
Thyroid Papillary Cancer | Poor prognosis | Poor prognosis |
Pancreatic Cancer | Poor prognosis | Poor prognosis |
Cholangiocarcinoma | Poor prognosis | Poor prognosis |
Cervical Cancer | Poor prognosis | Poor prognosis |
Interestingly, the prognostic value of CCND1 overexpression in breast cancer remains controversial, with some studies reporting it as a good prognostic marker [63-66] while others suggest it indicates poor prognosis . Research by Kyomoto et al. and Miyamoto et al. found that CCND1 amplification is a more effective and reliable prognostic marker than protein overexpression in head and neck squamous cell carcinoma .
To properly evaluate prognostic significance, researchers should:
Use multivariate analysis to control for confounding factors
Conduct survival analyses using Kaplan-Meier curves and log-rank tests
Ensure adequate sample sizes and follow-up periods
Consider subtypes within cancer categories (e.g., ER status in breast cancer)
Recent proteomic analyses have unexpectedly revealed that CCND1 interacts with a network of DNA repair proteins, most notably RAD51, a recombinase essential for homologous recombination (HR) :
Direct binding evidence: Experiments using purified recombinant proteins demonstrated that cyclin D1 directly binds to RAD51, with the N-terminus of cyclin D1 and C-terminus of RAD51 mediating this interaction .
Radiation-induced interaction: The physical interaction between endogenous cyclin D1 and RAD51 proteins intensifies with increased radiation dose, suggesting a functional role in the DNA damage response .
BRCA2-dependent recruitment: Like RAD51, cyclin D1 is recruited to DNA damage sites in a BRCA2-dependent fashion, placing it physically at locations where DNA repair occurs .
Researchers can investigate this interaction through:
Co-immunoprecipitation assays with endogenous proteins
GST pull-down assays with recombinant proteins to map interaction domains
Immunofluorescence microscopy to visualize co-localization at DNA damage sites
Proximity ligation assays to detect protein interactions in situ
CCND1 plays a critical role in homologous recombination (HR)-mediated DNA repair :
Functional impact on HR: Depletion of cyclin D1 in cancer cell lines (HeLa and H2009) significantly reduces HR efficiency, and this effect can be rescued by re-expression of siRNA-resistant cyclin D1 .
Mechanism of action: Cyclin D1 influences HR by affecting RAD51 recruitment to damaged DNA. Reduction of cyclin D1 levels impairs this recruitment process, thereby impeding HR-mediated DNA repair .
Therapeutic implications: Cyclin D1-depleted cells show increased sensitivity to PARP inhibitors, consistent with the established vulnerability of HR-deficient cells to these agents .
To experimentally measure HR efficiency, researchers can use:
DR-eGFP reporter system: This assay uses a GFP reporter construct containing an I-SceI endonuclease site to measure HR activity following DNA double-strand breaks .
RAD51 foci formation assays: Immunofluorescence detection of RAD51 foci formation after irradiation provides a measure of successful recruitment of repair factors.
Comet assay: To measure DNA damage resolution over time following treatment with genotoxic agents.
Cell survival assays: Colony formation assays following DNA damage or PARP inhibitor treatment can assess functional outcomes of HR deficiency.
The relationship between CCND1 and PARP inhibitor sensitivity has important implications for cancer therapy :
Sensitization effect: Experimental evidence demonstrates that cyclin D1-depletion sensitizes cancer cells to treatment with poly (ADP-ribose) polymerase (PARP)-inhibitors .
Mechanistic basis: This sensitivity stems from cyclin D1's role in homologous recombination (HR). Depletion of cyclin D1 impairs HR efficiency, creating a synthetic lethal interaction with PARP inhibition, similar to that observed in BRCA-deficient cells .
Therapeutic potential: This relationship suggests that tumors with low cyclin D1 expression might be particularly vulnerable to PARP inhibitor therapy, while conversely, high cyclin D1 expression might confer resistance.
Researchers investigating this relationship should consider:
Cell viability assays (MTT, CellTiter-Glo) to measure dose-dependent responses to PARP inhibitors
Combination index analysis to quantify synergy between cyclin D1 inhibition and PARP inhibitors
Xenograft models to assess in vivo responses to combination therapies
Analysis of patient-derived samples to correlate cyclin D1 expression with clinical responses to PARP inhibitors
Advanced methodologies for studying CCND1 protein interactions include:
Proteomic screening approaches: Double immunoaffinity purification coupled with liquid chromatography and high-throughput mass spectrometry (LC-MS/MS) has successfully identified 132 high-confidence CCND1-interacting proteins, revealing previously unknown functions like its role in DNA repair .
Proximity-based labeling methods: BioID or APEX2 fusion proteins can be used to identify proteins in close proximity to CCND1 in living cells, potentially revealing transient or weak interactions.
FRET/BRET analysis: These techniques allow real-time monitoring of protein-protein interactions in living cells and can detect conformational changes upon complex formation.
Cryo-electron microscopy: This approach can reveal the structural details of CCND1 complexes with CDK4/6 or DNA repair proteins like RAD51.
When designing interaction studies, researchers should:
Include appropriate controls to distinguish specific from non-specific interactions
Validate key interactions through reciprocal co-immunoprecipitation
Consider the impact of cellular context (cell type, cell cycle phase, etc.)
Determine the functional significance of interactions through mutational analysis
Distinguishing and targeting specific CCND1 isoforms presents methodological challenges that can be addressed through several approaches:
Isoform-specific antibodies: Develop antibodies targeting the unique C-terminal region of CCND1b to differentiate it from CCND1a in western blotting, immunoprecipitation, or immunohistochemistry.
RT-PCR with isoform-specific primers: Design primers spanning the alternative splice junction to selectively amplify each isoform for quantitative expression analysis.
Isoform-selective knockdown: Design siRNAs or antisense oligonucleotides targeting the unique regions of each isoform. Research has shown that antisense oligonucleotides can effectively target tumors carrying CCND1b .
CRISPR-based approaches: Use CRISPR/Cas9 to:
Create isoform-specific knockout cell lines by targeting unique exons
Introduce mutations at the G870A splice site to alter isoform ratios
Develop CRISPR interference (CRISPRi) constructs targeting isoform-specific promoter elements or regulatory regions
When employing these approaches, researchers should:
Validate isoform specificity through multiple complementary techniques
Consider compensatory mechanisms that may arise after targeting one isoform
Assess the impact on both cell cycle regulation and DNA repair functions
Researchers frequently encounter contradictory findings regarding CCND1 function, particularly related to its prognostic significance and isoform-specific activities . To properly interpret these contradictions:
Consider tissue-specific contexts: CCND1's function varies across tissue types. For example, CCND1 overexpression shows contradictory prognostic associations in breast cancer but consistently indicates poor prognosis in head and neck cancers .
Account for isoform differences: CCND1a and CCND1b have distinct and sometimes opposing functions. While CCND1a accelerates cell proliferation by promoting cell cycle progression, CCND1b may inhibit cell cycle progression in some contexts .
Evaluate experimental models: Different cell lines and experimental systems may yield conflicting results due to their unique genetic backgrounds and signaling pathway configurations.
Assess methodological differences: Variations in techniques used to measure CCND1 (e.g., antibodies recognizing different epitopes) may contribute to apparently contradictory findings.
Methodological approaches to resolve contradictions include:
Meta-analysis of multiple studies to identify patterns and sources of variation
Side-by-side comparison of different cell models under identical experimental conditions
Comprehensive characterization of the genetic and epigenetic background of experimental models
Detailed isoform-specific expression analysis in the experimental system under study
Accurate quantification of CCND1 expression in patient samples requires careful methodological considerations:
Distinguish amplification from overexpression: Research indicates that although CCND1 amplification correlates with overexpression, many cases of overexpression occur without gene amplification . Therefore, multiple techniques should be employed:
FISH or comparative genomic hybridization for gene amplification
RT-qPCR for mRNA expression
Immunohistochemistry or Western blotting for protein expression
Standardize scoring methods: When using immunohistochemistry, implement standardized scoring systems (e.g., H-score, Allred score) to ensure consistent interpretation across different studies.
Consider heterogeneity: Tumor heterogeneity can affect CCND1 expression assessment. Multiple regions of the tumor should be sampled when possible.
Distinguish between isoforms: When relevant, use isoform-specific methods (RT-PCR with isoform-specific primers or antibodies that distinguish CCND1a from CCND1b).
Include appropriate controls: Use positive and negative control tissues with established CCND1 expression patterns to validate assay performance.
Studies have shown that CCND1 amplification may be a more reliable prognostic marker than protein overexpression in certain cancers, highlighting the importance of comprehensive assessment approaches .
Based on current research, several promising strategies are emerging for targeting CCND1 dysregulation in cancer therapy:
Isoform-specific targeting: Given the distinct functions of CCND1a and CCND1b, developing therapeutics that selectively target the oncogenic CCND1b isoform while sparing CCND1a could provide more precise intervention with fewer side effects .
CDK4/6 inhibition combined with CCND1 targeting: Current CDK4/6 inhibitors (palbociclib, ribociclib, abemaciclib) might be more effective when combined with direct CCND1 targeting strategies.
Exploiting synthetic lethality: The role of CCND1 in homologous recombination suggests potential synthetic lethal approaches. Cyclin D1-depleted cells show increased sensitivity to PARP inhibitors , indicating a therapeutic opportunity for combination strategies.
RNA-based therapeutics: Antisense oligonucleotides or small interfering RNAs targeting CCND1b have shown promise in experimental systems and represent an emerging therapeutic approach with increasing clinical feasibility.
Targeting CCND1 regulators: Modulating the expression of trans-acting factors that regulate CCND1 splicing could alter the ratio of CCND1a to CCND1b, potentially providing therapeutic benefit.
Research approaches to evaluate these strategies include:
High-throughput drug screening in cell lines with defined CCND1 status
Patient-derived xenograft models to assess therapeutic efficacy in more clinically relevant systems
CRISPR-based genetic screens to identify synthetic lethal interactions with CCND1 alterations
Combination therapy evaluations in preclinical models
Single-cell technologies offer unprecedented opportunities to uncover new aspects of CCND1 biology:
Heterogeneity in expression and function: Single-cell RNA sequencing can reveal cell-to-cell variability in CCND1 isoform expression and correlate this with cell cycle state, providing insights into how differential expression affects cellular outcomes.
Cell cycle dynamics: Single-cell time-lapse imaging combined with CCND1 fluorescent reporters can capture real-time dynamics of CCND1 expression, localization, and degradation throughout the cell cycle.
Spatial context: Spatial transcriptomics and multiplexed protein imaging can map CCND1 expression patterns within tissues, potentially revealing microenvironmental influences on CCND1 regulation.
Rare cell populations: Single-cell approaches can identify and characterize rare subpopulations of cells with unique CCND1 expression patterns that might drive cancer progression or treatment resistance.
Response to perturbation: Single-cell analysis following genetic or pharmacological perturbation can reveal how individual cells adjust CCND1 expression and function in response to specific stimuli.
Methodological considerations for single-cell CCND1 studies include:
Careful sample preparation to preserve cell integrity and native state
Inclusion of cell cycle markers to properly interpret CCND1 variation
Development of computational methods to integrate multi-modal single-cell data
Validation of single-cell findings in appropriate model systems