CCNG1 Human

Cyclin G1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to CCNG1 Human

CCNG1 (Cyclin G1) Human is a recombinant protein produced for research applications, derived from the CCNG1 gene (NCBI Gene: 900). This non-canonical cyclin plays dual roles in cell cycle regulation and oncogenesis, acting as a critical node in the Cyclin G1/p53/Mdm2 axis . Unlike canonical cyclins, CCNG1 lacks the PEST protein-destabilizing sequence, enhancing its stability .

Molecular Composition

  • Recombinant Production: CCNG1 Human Recombinant is synthesized in E. coli as a 315-amino acid polypeptide (36.2 kDa), fused with an N-terminal His-tag for purification .

  • Domains: Contains a cyclin box but lacks CDK-binding residues, enabling unique interactions with PP2A phosphatase and Mdm2 .

Biological Functions

  • Cell Cycle Regulation: Governs G0/G1 transition and G2/M arrest during DNA damage .

  • Oncogenic Role: Stabilizes c-Myc and Mdm2, suppressing p53 tumor suppressor activity to promote cancer cell survival .

CCNG1 in Cancer

Cancer TypeExpression LevelClinical Correlation
Melanoma↑↑ (Metastatic)Linked to chemo-resistance and poor prognosis
Breast Cancer35–95% nuclear stainingAssociated with HER2+ subtypes
Esophageal Cancer↑ in radioresistant cellsDrives Wnt/β-catenin pathway activation
Ovarian CancerUpregulated in 80% of HGSOCCorrelates with mutant p53

Key Pathways

  • Cyclin G1/Mdm2/p53 Axis: CCNG1 binds PP2A to dephosphorylate Mdm2, accelerating p53 degradation and enabling unchecked proliferation .

  • c-Myc Stabilization: CCNG1/CDK5 phosphorylates c-Myc, enhancing DNA repair gene transcription despite genomic instability .

Biomarker Potential

  • CCNG1 mRNA and protein levels are elevated in 14+ cancer types compared to normal tissues (p = 0.006–1.8E-58) .

  • Positively correlates with Ki-67 (r = 0.937, p < 0.00001), indicating proliferative activity .

Therapeutic Strategies

  • DeltaRex-G: A CCNG1 inhibitor showing 9–12 year survivorship in metastatic cancers .

  • siRNA/Peptide Inhibitors: Targeting CCNG1’s α3/α5 helices induces apoptosis in preclinical models .

Future Directions

  • Phase II Trials: Basket studies to validate CCNG1 as a predictive biomarker for inhibitor therapies .

  • Mechanistic Studies: Elucidate CCNG1’s role in cancer stem cell competence and EMT .

Product Specs

Introduction
Cyclin-G1 (CCNG1), a member of the cyclin family, contains the cyclin box and is believed to play a role in growth regulation. It is linked to G2/M phase arrest in response to DNA damage. CCNG1 might act as an intermediary through which p53 exerts its inhibitory effect on cellular proliferation. Interestingly, CCNG1 lacks the protein destabilizing (PEST) sequence found in other members of the cyclin family.
Description
Recombinant human CCNG1, expressed in E. coli, is a single, non-glycosylated polypeptide chain comprising 315 amino acids (specifically, amino acids 1 to 295). It has a molecular weight of 36.2 kDa. For purification purposes, a 20 amino acid His-tag is fused to the N-terminus of CCNG1, and purification is achieved using proprietary chromatographic techniques.
Physical Appearance
The product is a clear, colorless solution that has been sterilized by filtration.
Formulation
The CCNG1 protein is supplied in a solution with a concentration of 0.25 mg/ml. The solution contains 20 mM Tris-HCl buffer (pH 8.0), 50% glycerol, 0.2 M NaCl, and 5 mM DTT.
Stability
For short-term storage (up to 2-4 weeks), the product should be kept at 4°C. For longer-term storage, it is recommended to freeze the product at -20°C. To ensure optimal stability during long-term storage, consider adding a carrier protein (0.1% HSA or BSA). It is important to avoid repeated cycles of freezing and thawing.
Purity
Analysis by SDS-PAGE indicates that the purity of the product is greater than 85%.
Synonyms
Cyclin-G1, Cyclin-G, CCNG1, CCNG, CYCG1.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MIEVLTTTDS QKLLHQLNAL LEQESRCQPK VCGLRLIESA HDNGLRMTAR LRDFEVKDLL SLTQFFGFDT ETFSLAVNLL DRFLSKMKVQ PKHLGCVGLS CFYLAVKSIE EERNVPLATD LIRISQYRFT VSDLMRMEKI VLEKVCWKVK ATTAFQFLQL YYSLLQENLP LERRNSINFE RLEAQLKACH CRIIFSKAKP SVLALSIIAL EIQAQKCVEL TEGIECLQKH SKINGRDLTF WQELVSKCLT EYSSNKCSKP NVQKLKWIVS GRTARQLKHS YYRITHLPTI PEMVP.

Q&A

What is CCNG1 and what is its role in cell cycle regulation?

CCNG1 is the gene encoding Cyclin G1, a member of the cyclin family initially identified with homology to C-SRC. It functions as a pivotal component of the cyclin G1/Mdm2/p53 axis and plays a strategic role in cell cycle checkpoint control . Unlike other cyclins that typically promote cell cycle progression through positive regulation of cyclin-dependent kinases (CDKs), Cyclin G1 primarily functions in a complex regulatory network involving p53, a key tumor suppressor protein. Cyclin G1 was first recognized as a p53-regulated transcript induced by DNA damage, establishing it as a cell cycle regulator with unique properties compared to other cyclin family members .

How has understanding of CCNG1 function evolved in cancer research?

The understanding of CCNG1 has evolved significantly from its initial characterization as a cell cycle regulator to recognition of its complex roles in cancer biology. Research has identified Cyclin G1 as a cell cycle regulator in multiple human tumor types, including cervical carcinoma, hepatocellular carcinoma, breast cancer, and lung carcinoma . Moreover, CCNG1 has emerged as a potential biomarker associated with recurrence and chemoresistance in hepatocellular carcinoma . Recent studies have linked CCNG1 amplification with significantly shorter post-surgical survival in patients with ovarian cancer, highlighting its prognostic significance . This evolution in understanding has positioned CCNG1 as not merely a cell cycle component but as a multifunctional regulator with significant implications for cancer progression and treatment response.

How does Cyclin G1 interact with the Mdm2/p53 axis?

Cyclin G1 functions as a critical regulatory component of the cyclin G1/Mdm2/p53 axis that controls cell cycle progression and checkpoint responses. The mechanism involves Cyclin G1's ability to modulate p53 function through interaction with Mdm2, which is a negative regulator of p53 . This interaction creates a regulatory feedback loop where:

  • Cyclin G1 can form complexes that influence the phosphorylation state of Mdm2

  • This modulation affects Mdm2's ability to target p53 for degradation

  • The resulting changes in p53 levels and activity influence cell cycle progression and apoptotic responses

The importance of this axis is underscored by research showing that this pathway provides a mechanistic basis for understanding the broad-spectrum anticancer activity observed with dominant-negative cyclin G1, which can trigger programmed cell death when introduced into cancer cells .

What signaling pathways interact with CCNG1 during cellular stress responses?

CCNG1 participates in multiple signaling networks during cellular stress, particularly DNA damage responses. As a p53-regulated transcript induced by DNA damage, Cyclin G1 functions in:

  • The DNA damage response pathway, where its expression is upregulated following genotoxic stress

  • Cell cycle checkpoint pathways, particularly in mitotic checkpoint control during taxane treatment

  • Pathways involving Protein Phosphatase 2A (PP2A), which affects the phosphorylation status of key cell cycle proteins

Research indicates that CCNG1 may interact with NF-κB signaling pathways, as suggested by studies examining TNIP1 (tumor necrosis factor α-induced protein 3-interacting protein 1) and CCNG1 knockdown effects on breast cancer cells .

What are effective methods for CCNG1 knockdown in experimental models?

Based on published research methodologies, effective CCNG1 knockdown can be achieved through several approaches:

RNA interference techniques:

  • Short hairpin RNAs (shRNAs) targeting CCNG1 inserted into vectors such as pLKO.1 plasmids have been successfully used for stable knockdown

  • siRNA-mediated knockdown using specific sequences can be employed for transient depletion, with protocols often involving double transfection (initial reverse transfection followed by forward transfection 24 hours later) to enhance knockdown efficiency

Transfection protocols:

  • For siRNA delivery, Dharmafect 1 has been utilized with standard protocols at concentrations of approximately 50-100nM

  • For stable knockdown, lentiviral transduction systems with plasmids such as pLKO.1 containing shRNAs co-transfected with packaging plasmids (psPAX2 and pMD2.G in a ratio of 4:3:1) have proven effective

Validation methods:

  • Quantitative RT-PCR using primers targeting CCNG1 mRNA and normalizing to housekeeping genes such as PBGD or GAPDH using the ΔΔCt methodology

  • Western blotting to confirm protein level reduction

How can researchers assess the functional impact of CCNG1 modulation in cancer cells?

Researchers can employ multiple complementary assays to assess the functional impact of CCNG1 modulation:

Cell proliferation assays:

  • MTT assays: Cells can be seeded in 96-well plates (typically 2,500 cells/well) and assessed at 0, 24, and 48-hour timepoints. The formazan product is quantified spectrophotometrically at 570nm

  • Colony formation assays: 1,000 cells with stable CCNG1 knockdown can be seeded in 6-well plates, cultured for seven days, then fixed with paraformaldehyde and stained with crystal violet for colony counting

Cell cycle and apoptosis analysis:

  • Flow cytometry for cell cycle distribution and apoptosis detection

  • Live-cell imaging to monitor mitotic progression using time-lapse microscopy, collecting images every 3-5 minutes over 48 hours to track cell division events

Cell viability measurements:

  • CellTiter-Blue Cell Viability Assay or similar fluorescence-based methods (excitation: 520nm, emission: 590nm) to quantify viable cells following CCNG1 modulation

How does CCNG1 expression influence chemotherapy response?

CCNG1 expression has significant implications for chemotherapy response, particularly for taxane-based treatments. Research indicates that:

  • CCNG1 amplification correlates with shorter post-surgical survival in ovarian cancer patients, suggesting a role in treatment resistance

  • Cyclin G1 regulates the outcome of taxane-induced mitotic arrest in cancer cells, potentially affecting sensitivity to this important class of chemotherapeutic agents

  • CCNG1 may serve as a biomarker for chemoresistance in hepatocellular carcinoma, indicating its potential utility in predicting treatment outcomes

The mechanism appears to involve CCNG1's role in mitotic checkpoint control, where it influences how cells respond to microtubule-targeting agents like taxanes. This suggests that modulation of CCNG1 expression or activity could potentially be used to enhance chemosensitivity in resistant tumors.

What is the relationship between CCNG1 and tumor suppressor pathways?

CCNG1 has a complex relationship with tumor suppressor pathways, particularly the p53 pathway:

  • While CCNG1 is identified as a p53-regulated transcript induced by DNA damage , it can also function to disable p53 function in certain contexts

  • Research has positioned Cyclin G1 in opposition to tumor suppressor proteins such as p53, pRb, p16INK4A, and p21WAF1, which are commonly dysregulated in cancer

  • Cyclin G1 appears to be involved in a regulatory mechanism that can potentially override p53 checkpoints and advance cell cycle progression

This bidirectional relationship with tumor suppressor pathways makes CCNG1 a particularly interesting target for cancer research, as it suggests potential for therapeutic intervention at this node of cell cycle control.

How do post-translational modifications regulate Cyclin G1 function?

Post-translational modifications (PTMs) of Cyclin G1 represent an area that requires further investigation, but current research suggests several important regulatory mechanisms:

  • Phosphorylation events likely play a crucial role in regulating Cyclin G1's interactions with binding partners such as Mdm2 and protein phosphatase 2A (PP2A)

  • The interaction between Cyclin G1 and protein phosphatase complexes suggests that dephosphorylation events mediated by these interactions constitute an important regulatory mechanism in the p53 pathway

  • The stability and turnover of Cyclin G1 protein may be regulated by ubiquitination pathways, similar to other cyclins, though the specific E3 ligases involved require further characterization

Understanding these modifications is critical for developing a complete picture of how Cyclin G1 function is dynamically regulated in different cellular contexts and in response to various stimuli.

What are the implications of CCNG1 in cancer stem cell biology?

The role of CCNG1 in cancer stem cell biology represents an emerging area of research with potential significance for understanding tumor initiation, progression, and treatment resistance:

  • Given CCNG1's role in cell cycle regulation and its association with chemoresistance, it may contribute to the characteristic therapy resistance of cancer stem cell populations

  • The involvement of CCNG1 in the p53 pathway, which is important for stem cell maintenance and differentiation, suggests potential roles in determining cancer stem cell fate decisions

  • Research examining the connection between CCNG1 expression and stemness markers in various cancer types could provide insights into whether CCNG1 contributes to the acquisition or maintenance of stem-like properties in cancer cells

This area represents a promising frontier for CCNG1 research, particularly in developing strategies to target therapy-resistant cancer stem cell populations.

What are the recommended cell models for studying CCNG1 function?

Based on the literature, several cell models have proven valuable for CCNG1 research:

Breast cancer models:

  • MCF-7 and T47D human breast cancer cell lines have been successfully used for CCNG1 knockdown studies and functional assessments

Other validated models:

  • HEK293T cells are useful for lentiviral production for stable CCNG1 knockdown experiments

  • Various cancer cell lines representing different tissue types (hepatocellular carcinoma, cervical carcinoma, lung carcinoma) where CCNG1 has demonstrated regulatory roles

When selecting a model system, researchers should consider:

  • Baseline CCNG1 expression levels

  • p53 status (wild-type vs. mutant)

  • Relevant genetic background for the specific research question

  • Growth characteristics and amenability to experimental manipulation

How can CCNG1 expression be accurately quantified in clinical samples?

Accurate quantification of CCNG1 expression in clinical samples requires robust and validated methodologies:

mRNA quantification:

  • Quantitative RT-PCR using validated primer sets (sequences available in the literature ) and appropriate reference genes

  • RNA-seq analysis with proper normalization procedures

  • In situ hybridization techniques for spatial expression analysis in tissue sections

Protein detection:

  • Immunohistochemistry (IHC) with validated antibodies and appropriate scoring systems

  • Western blotting for semi-quantitative protein assessment

  • Protein arrays or mass spectrometry-based proteomics approaches for broader protein interaction studies

For clinical application, standardized protocols should be developed with:

  • Defined sample collection and processing procedures

  • Validated cutoff values for categorizing expression levels

  • Quality control measures to ensure reproducibility

  • Correlation with known clinical parameters to establish predictive or prognostic value

Product Science Overview

Introduction

Cyclin G1 is a member of the cyclin family, which plays a crucial role in regulating the cell cycle. Cyclins are proteins that control the progression of cells through the cell cycle by activating cyclin-dependent kinases (CDKs). Cyclin G1, in particular, is involved in the regulation of the G1 phase of the cell cycle, which is the first phase in the cell cycle that leads to cell division.

Structure and Function

Cyclin G1 is characterized by its cyclical expression pattern, which is tightly regulated during the cell cycle. It forms complexes with CDKs, particularly CDK2 and CDK4, to control the transition from the G1 phase to the S phase, where DNA replication occurs. The activity of Cyclin G1 is essential for the proper timing of cell cycle events and ensuring that cells only divide when they are ready.

Human Recombinant Cyclin G1

Human recombinant Cyclin G1 is a form of the protein that is produced using recombinant DNA technology. This involves inserting the gene that encodes Cyclin G1 into a host organism, such as bacteria or yeast, which then produces the protein. This method allows for the production of large quantities of Cyclin G1 for research and therapeutic purposes.

Role in Cancer Research

Cyclin G1 has been extensively studied in the context of cancer research. Abnormal regulation of Cyclin G1 and its associated CDKs can lead to uncontrolled cell proliferation, a hallmark of cancer. Understanding the mechanisms by which Cyclin G1 regulates the cell cycle can provide insights into potential therapeutic targets for cancer treatment.

Applications

Human recombinant Cyclin G1 is used in various research applications, including:

  • Cell Cycle Studies: Researchers use Cyclin G1 to study the regulation of the cell cycle and identify potential points of intervention for cancer therapy.
  • Drug Development: Cyclin G1 is used in screening assays to identify compounds that can modulate its activity, which may lead to the development of new cancer treatments.
  • Biotechnology: The production of recombinant Cyclin G1 is an example of how recombinant DNA technology can be used to produce proteins for research and therapeutic use.

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.