GINS2 (also known as Psf2) is a core component of the eukaryotic replicative helicase, facilitating DNA replication initiation and cell cycle progression . The GINS complex (Sld5, Psf1, Psf2, Psf3) ensures genomic stability during S-phase, making GINS2 critical for cellular proliferation . Dysregulation of GINS2 is implicated in tumorigenesis via pathways like MAPK/ERK, p53, and DNA damage response (DDR) .
Cell Cycle Arrest: GINS2 knockdown induced G1-phase arrest by downregulating CDK4, CDK6, and Cyclin D1 .
Apoptosis Regulation: Silencing GINS2 increased pro-apoptotic Bax and decreased anti-apoptotic Bcl-2, promoting apoptosis .
MAPK/ERK Pathway: GINS2 interference reduced MEK, p-MEK, ERK, and p-ERK levels, suppressing tumor growth in xenograft models .
| Protein | Expression Change (GINS2 Knockdown) | Pathway Impact |
|---|---|---|
| Bax | ↑ 2.5-fold | Pro-apoptotic |
| Bcl-2 | ↓ 60% | Anti-apoptotic |
| CDK4/CDK6 | ↓ 70% | Cell cycle arrest |
| p-ERK | ↓ 55% | MAPK/ERK suppression |
PTP4A1/p53 Axis: GINS2 knockdown reduced proliferation and increased apoptosis via PTP4A1 downregulation, which restored p53 activity .
Rescue Experiments: Overexpressing PTP4A1 reversed shRNA-GINS2 effects on cyclin D1, Bcl2, and Bax .
Cell Cycle Blockade: GINS2 inhibition caused G2/M-phase arrest by elevating ATM, CHK2, and p53, indicating DDR activation .
Proliferation Impact: GINS2 overexpression accelerated acute promyelocytic leukemia (APL) cell growth .
Chemoresistance: GINS2 promoted temozolomide (TMZ) resistance via the EGR1/ECT2 axis, enhancing DDR and stemness .
Therapeutic Synergy: Combining TMZ with a GINS2 inhibitor (identified via CMap screening) suppressed glioma proliferation synergistically .
In hepatocellular carcinoma (HCC), GINS2 expression correlated with immune infiltration:
Positive Associations: Infiltrating B cells (r=0.386, P<0.001), CD8+ T cells (r=0.221, P<0.001), and macrophages (r=0.12, P=0.026) .
Key Immune Markers: GINS2 co-expressed with CDK1 (r=0.75, P<0.001) and CCNB1 (r=0.742, P<0.001), linking cell cycle progression to immune evasion .
| Immune Cell Type | Marker Gene | Correlation (r) | P-Value |
|---|---|---|---|
| CD8+ T cells | CD8A | 0.268 | 4.37E-07 |
| M1 Macrophages | IRF5 | 0.26 | 3.50E-07 |
| Activated T cells | CD69 | 0.159 | 0.003 |
GINS2 (Go-Ichi-Ni-San 2), also known as PSF2 (Partner of Sld Five 2), is a critical component of the GINS complex that plays an essential role in DNA replication. In humans, GINS2 is a 185 amino acid protein with a molecular weight of approximately 21.4 kDa . It functions primarily in the nucleus and chromosomes as part of the CDC45-MCM-GINS (CMG) helicase complex, the molecular machine that unwinds template DNA during replication and around which the replisome is built .
The biological significance of GINS2 lies in its role in both the initiation of DNA replication and the progression of DNA replication forks . Without proper GINS2 function, cells cannot effectively replicate their DNA, which impacts cell cycle progression and genomic stability. The collaboration of GINS2 with other proteins in the replication complex ensures successful DNA unwinding and synthesis, demonstrating its critical role in cell proliferation and genomic stability .
GINS2 antibodies have been validated for multiple research applications, with Western Blot (WB) being the most widely used and extensively validated technique. According to published literature and product data, GINS2 antibodies have been successfully employed in:
Western Blot (WB): Most commercial antibodies show strong validation for detecting GINS2 at approximately 25 kDa (observed molecular weight)
Immunohistochemistry (IHC-P): Used for detection in paraffin-embedded tissue sections
Immunofluorescence (IF/ICC): Validated for cellular localization studies
ELISA: Some antibodies are suitable for enzyme-linked immunosorbent assays
Research publications have predominantly cited Western Blot applications, with at least 11 publications specifically using this method for GINS2 detection .
GINS2 expression has been documented in multiple cell types and tissues, with notable patterns in both normal and pathological samples:
Cancer cell lines with confirmed GINS2 expression:
Tissue expression patterns:
High expression in epithelial ovarian cancer (EOC) tissues (58.33% of samples) compared to normal ovarian tissues (16.67%)
Weak expression in normal ovarian tissue, with some signals in blood vessels
Altered expression in peripheral blood of patients with intervertebral disk degeneration
This expression profile makes GINS2 a valuable research target for understanding both normal cellular processes and pathological conditions, particularly in oncology research.
Western Blot Protocol:
Expected molecular weight: 21 kDa (calculated), typically observed at 25 kDa
Sample preparation: Total protein extraction from cells/tissues with standard lysis buffer containing protease inhibitors
Loading control: β-actin is commonly used as internal reference
Detection method: ECL reagent (Thermo Scientific Pierce) has been successfully used
Immunohistochemistry Protocol:
Antigen retrieval: TE buffer pH 9.0 (alternatively, citrate buffer pH 6.0)
Positive control tissues: Pancreatic cancer tissue has shown reliable staining
Visualization: DAB chromogen with hematoxylin counterstain
Immunofluorescence/ICC Protocol:
Cell fixation: 4% paraformaldehyde followed by permeabilization with 0.1% Triton X-100
Blocking: 5% BSA in PBS
Nuclear counterstain: DAPI
Flow Cytometry Protocol:
Recommended amount: 0.25 μg per 10^6 cells in 100 μl suspension
Permeabilization required for intracellular staining
Validating antibody specificity is crucial for reliable results. For GINS2 antibodies, consider these validation approaches:
Genetic validation:
Expression validation:
Peptide competition assay:
Pre-incubate antibody with immunizing peptide/protein
Expected result: Signal abolishment or significant reduction
Multiple antibody validation:
Use antibodies targeting different epitopes of GINS2
Expected result: Similar detection pattern across antibodies
Molecular weight verification:
Several factors can significantly impact GINS2 antibody performance in Western blot applications:
Sample preparation:
Complete protein denaturation is essential
Use fresh samples when possible
Include protease inhibitors in lysis buffer to prevent degradation
Antibody dilution optimization:
Blocking conditions:
Typically 5% non-fat milk or BSA in TBST
Some antibodies may perform better with specific blocking agents
Incubation conditions:
Detection method sensitivity:
ECL reagents of appropriate sensitivity for the expected expression level
Exposure time optimization to avoid saturation
Post-translational modifications:
GINS2 expression alterations have been documented in multiple cancer types, with antibody-based detection methods playing a crucial role in these discoveries:
Cancer types with altered GINS2 expression:
Epithelial ovarian cancer (EOC): 58.33% of EOC tissues showed high GINS2 expression compared to 16.67% in normal ovarian tissues
Breast cancer: GINS2 transcript highly expressed, with expression levels correlating with histological grade (17.2% in grade 1, 50% in grade 2, and 77.1% in grade 3)
Lung adenocarcinoma: Identified as a differentially expressed gene (high expression) in stage II
Gastric adenocarcinoma, pancreatic cancer: Carcinogenic role reported
Antibody applications in cancer research:
Tissue expression profiling: IHC with GINS2 antibodies can reveal expression patterns across different tumor grades and stages
Prognostic marker evaluation: Correlating GINS2 expression with patient outcomes
Mechanistic studies: When combined with functional assays following GINS2 knockdown or overexpression
Therapeutic response monitoring: Evaluating GINS2 expression changes following treatment
Research has demonstrated that manipulating GINS2 expression levels (through knockdown or overexpression) affects cancer cell behavior, suggesting its potential as a therapeutic target .
An intriguing and unexpected role for GINS2 was discovered in intervertebral disk degeneration (IDD), where its expression pattern differs from that observed in cancer:
Key findings on GINS2 in IDD:
GINS2 is significantly downregulated in the peripheral blood of patients with intervertebral disk degeneration
This was initially discovered through bioinformatics analysis of the GEO database (GSE124272) and subsequently validated in clinical samples
ROC curve analysis yielded a high AUC of 0.8261 (95% CI = 0.7051-0.9472), suggesting GINS2 could serve as a potential diagnostic biomarker for IDD
Functional significance in nucleus pulposus cells:
Overexpression of GINS2 increased proliferation, migration, and invasion of nucleus pulposus (NP) cells
GINS2 overexpression decreased apoptotic properties of NP cells
These findings suggest GINS2 might be a potential therapeutic target for IDD
The discovery process involved:
Initial bioinformatics screening of differentially expressed genes
Validation in peripheral blood samples from 30 IDD patients and 30 healthy participants
Functional studies in NP cells using GINS2 overexpression plasmids
Evaluation of cellular behaviors using proliferation, migration, invasion, and apoptosis assays
To investigate correlations between GINS2 expression and clinical outcomes, researchers should consider the following methodological approaches:
Tissue microarray (TMA) analysis:
Use validated GINS2 antibodies (1:50-1:500 dilution for IHC)
Score staining intensity and percentage of positive cells
Large patient cohorts with complete clinical data are essential
Example scoring system: High expression defined as positive staining in >50% of cells with moderate-to-strong intensity
Peripheral blood analysis:
RNA extraction followed by RT-qPCR for GINS2 mRNA quantification
Protein analysis using ELISA or Western blot with GINS2 antibodies
Standardized collection protocols to minimize pre-analytical variables
Statistical analysis approaches:
Receiver operating characteristic (ROC) curves to assess diagnostic potential (as demonstrated for IDD with AUC = 0.8261)
Kaplan-Meier survival analysis stratified by GINS2 expression levels
Cox proportional hazards regression for multivariate analysis
Correlation analysis with established clinical parameters
Longitudinal sample collection:
Baseline and follow-up samples to track expression changes over disease progression
Correlation with treatment response and disease recurrence
Multi-omics integration:
GINS2 undergoes multiple post-translational modifications (PTMs) that can significantly impact antibody binding and experimental outcomes:
Documented PTMs of GINS2:
| Site | PTM Type | Source |
|---|---|---|
| M1 | Acetylation | Uniprot |
| K70 | Ubiquitination | Uniprot |
| K73 | Ubiquitination | Uniprot |
| K80 | Ubiquitination | Uniprot |
| S104 | Phosphorylation | Uniprot |
| K109 | Ubiquitination | Uniprot |
| K118 | Ubiquitination | Uniprot |
| T158 | Phosphorylation | Uniprot |
| S179 | Phosphorylation | Uniprot |
Implications for antibody selection:
Epitope consideration: Select antibodies whose epitopes do not include or overlap with known PTM sites if detecting total GINS2 is the goal
Modification-specific antibodies: For studying specific PTMs, choose antibodies that specifically recognize phosphorylated or ubiquitinated forms
Multiple antibody approach: Use antibodies targeting different regions to ensure comprehensive detection
Experimental design considerations:
Sample preparation:
Include phosphatase inhibitors when studying phosphorylated forms
Include deubiquitinating enzyme inhibitors when studying ubiquitinated forms
Consider lambda phosphatase treatment to remove phosphorylation if it interferes with detection
Migration pattern awareness:
Cell cycle considerations:
GINS2 phosphorylation may vary throughout the cell cycle
Cell synchronization might be necessary for studying specific modifications
Both polyclonal and monoclonal GINS2 antibodies are available for research, each with distinct characteristics that make them suitable for different applications:
Polyclonal GINS2 antibodies:
Target multiple epitopes across the GINS2 protein
Examples: Proteintech 16247-1-AP, Abcam ab197123, Affinity Biosciences DF9451
Advantages:
Higher sensitivity due to recognition of multiple epitopes
More robust to minor protein denaturation or fixation effects
Better for detection of low abundance targets
Best used for:
Monoclonal GINS2 antibodies:
Generated in mice or rabbits
Target a single epitope on GINS2
Applications: Typically more specific for particular applications
Advantages:
Consistent lot-to-lot reproducibility
Higher specificity for a single epitope
Lower background in some applications
Best used for:
Studies requiring high reproducibility
Specific epitope targeting
Applications where background is problematic
Selection guide based on research needs:
For novel research areas or initial characterization: Start with polyclonal antibodies for broader detection capabilities
For reproducible, long-term studies: Consider monoclonal antibodies once the detection parameters are established
For targeting specific forms/regions: Select antibodies with appropriate epitope specificity
For cross-species studies: Check validated reactivity (human and mouse are most commonly validated)
When encountering inconsistent results with GINS2 antibodies across different experimental systems, consider this systematic troubleshooting approach:
1. Antibody validation issues:
Verify antibody specificity using positive and negative controls
Consider using GINS2 knockdown or knockout systems as negative controls
Test multiple antibodies targeting different epitopes
Check lot-to-lot variation by requesting Certificate of Analysis
2. Sample preparation factors:
Protein extraction method: Different buffers may affect epitope accessibility
Fixation conditions (for IHC/IF): Overfixation can mask epitopes
Protein denaturation (for WB): Ensure complete denaturation with heat and reducing agents
Sample storage: Degradation due to improper storage or freeze-thaw cycles
3. Technical parameters to optimize:
Antibody dilution: Titrate across a wider range than recommended
Incubation conditions: Time and temperature adjustments
Blocking reagents: Test alternative blocking solutions (BSA vs. milk)
Detection systems: Try more sensitive detection methods
4. Biological variables to consider:
Cell cycle stage: GINS2 expression and localization may vary across the cell cycle
Cell confluence: Growth conditions can affect expression levels
Post-translational modifications: Different conditions may alter modification patterns
Species differences: Check if the antibody is validated for your species of interest
5. Systematic optimization approach:
Change only one variable at a time
Document all conditions thoroughly
Include internal controls in each experiment
Consider positive controls from published studies (e.g., Jurkat, HeLa, or SKOV-3 cells)
6. Application-specific troubleshooting:
For WB: Try different membrane types, transfer conditions, or detection systems
For IHC: Test multiple antigen retrieval methods
For IF: Adjust permeabilization conditions
For Flow cytometry: Optimize fixation and permeabilization protocols
GINS2 antibodies can serve as powerful tools for dissecting the protein's function within the DNA replication complex through several advanced approaches:
Co-immunoprecipitation (Co-IP) studies:
Use GINS2 antibodies to pull down the entire GINS complex and associated proteins
Can identify interaction partners in the CDC45-MCM-GINS (CMG) helicase complex
Western blot analysis of immunoprecipitates can confirm known interactions or identify novel binding partners
Mass spectrometry analysis of immunoprecipitates can provide unbiased identification of the complete interactome
Chromatin immunoprecipitation (ChIP) assays:
Use GINS2 antibodies to identify genomic regions where the GINS complex is bound
Can map GINS2 localization at replication origins and forks
ChIP-seq approaches provide genome-wide binding profiles
Time-course experiments can track GINS2 movement during S phase
Proximity ligation assays (PLA):
Visualize protein-protein interactions in situ
Combine GINS2 antibodies with antibodies against other replication factors
Can reveal spatial and temporal dynamics of interactions
Immunofluorescence co-localization:
Use GINS2 antibodies alongside markers for replication forks (e.g., PCNA, RPA)
Can track assembly and disassembly of replication complexes
Live-cell imaging with fluorescently tagged antibody fragments can monitor dynamics
Cell cycle synchronization experiments:
Track GINS2 expression, localization, and complex formation throughout the cell cycle
Compare normal cells with cancer cells where replication may be dysregulated
Combine with inhibitors of cell cycle checkpoints to dissect regulatory mechanisms
Emerging research suggests GINS2 may have significant potential as a therapeutic target in various diseases, with antibodies playing a crucial role in advancing this field:
Current therapeutic potential findings:
In cancer therapy:
In intervertebral disk degeneration (IDD):
Antibody contributions to therapeutic development:
Target validation:
Use of GINS2 antibodies in tissue microarrays to confirm expression in patient samples
IHC analysis to correlate expression with disease progression and patient outcomes
Western blot and qPCR validation of knockdown efficiency in functional studies
Mechanism elucidation:
Pathway analysis using phospho-specific antibodies to determine downstream effectors
Co-IP studies to identify protein-protein interactions that could be therapeutically targeted
Subcellular localization studies to understand compartment-specific functions
Therapeutic antibody development:
While GINS2's nuclear localization makes it challenging for direct antibody targeting, antibodies can guide development of other therapeutic modalities
Antibody-based screening assays for small molecule inhibitors
Validation of target engagement in drug development
Biomarker applications:
Future research directions:
Development of cell-penetrating antibodies or antibody fragments targeting GINS2
Combining GINS2 targeting with standard therapies
Exploration of synthetic lethality approaches in cancers with high GINS2 expression
Understanding the differential expression of GINS2 across various physiological and pathological contexts can provide valuable insights into its biology and potential as a therapeutic target:
GINS2 in normal development and homeostasis:
Expression in actively proliferating tissues due to its role in DNA replication
Present in embryonic tissues with high replicative potential
Low or undetectable levels in most adult differentiated tissues
Detectable in adult tissues with high cellular turnover (bone marrow, intestinal epithelium)
Conserved across species, with orthologs reported in mouse, rat, bovine, frog, zebrafish, chimpanzee, and chicken
GINS2 in tissue regeneration and repair:
Potential upregulation during wound healing and tissue regeneration
In nucleus pulposus cells, GINS2 overexpression promotes proliferation, suggesting a role in tissue repair mechanisms
Likely activated during liver regeneration due to high replicative demands
GINS2 in pathological states:
Cancer:
Intervertebral disk degeneration:
Other degenerative conditions:
Potential dysregulation in conditions with aberrant cellular proliferation
May be involved in fibrotic disorders characterized by excessive cell proliferation
Experimental approaches to study differential expression:
Comparative IHC studies:
Use GINS2 antibodies on tissue microarrays containing normal, regenerating, and pathological samples
Quantitative scoring of expression levels and subcellular localization
Single-cell analysis:
Combine GINS2 antibodies with cell-type-specific markers
Flow cytometry or mass cytometry (CyTOF) for quantitative analysis
Single-cell RNA-seq with protein validation using antibodies
Developmental time-course studies:
Track GINS2 expression during embryonic development and postnatal growth
Compare with expression during tissue regeneration models
3D organoid cultures:
Study GINS2 expression and function in developing organoids
Compare normal versus disease-mimicking conditions
This comprehensive understanding of context-dependent GINS2 expression can guide more precise therapeutic targeting and biomarker development strategies.