The LIG1 Antibody is primarily used in research to study DNA replication, repair pathways, and cancer biology. Key applications include:
Western Blot (WB): To quantify LIG1 protein expression in cell lysates or tissue samples .
Immunoprecipitation (IP): To isolate LIG1 protein complexes for functional studies .
Immunohistochemistry (IHC): To localize LIG1 in tumor tissues or normal cells .
Flow Cytometry (FC): To analyze LIG1 expression in circulating cells or cancer cell populations .
Commercial Antibodies:
A comparison of widely used LIG1 antibodies is provided in Table 1.
| Antibody Vendor | Clone/Type | Applications | Reactivity | Citations |
|---|---|---|---|---|
| Proteintech | 67840-1-Ig | WB, CoIP, ELISA | Human, Mouse, Rat | 1 publication |
| Novus Biologicals | NB100-119 | WB, ICC, IP, RA | Human, Mouse | 27 references |
| antibodies-online | ABIN1526063 | WB, ICC, IP, IHC, FC | Human, Mouse | 12 references |
| Acris Antibodies GmbH | AM26565AF-N | WB, ICC, IP, IHC, FC | Human, Mouse | 8 references |
LIG1 is essential for sealing DNA nicks during replication and base excision repair (BER) . Its overexpression has been linked to aggressive cancer phenotypes:
Bladder Cancer: High LIG1 levels correlate with poor prognosis, tumor invasion, and resistance to immunotherapies (e.g., anti-PD-1/L1) .
Ovarian Cancer: LIG1 depletion sensitizes cells to platinum-based chemotherapy, reversing drug resistance .
Prostate Cancer: Combined LIG1 knockout and PARP inhibition triggers apoptosis in cancer cells .
Epithelial-to-Mesenchymal Transition (EMT): LIG1 regulates EMT pathways, promoting cancer metastasis .
Immune Evasion: High LIG1 expression suppresses tumor immune infiltration, limiting therapeutic responses .
LIG1 antibodies have also been implicated in autoimmune encephalitis, where anti-LGI1 antibodies disrupt synaptic function, causing seizures and cognitive decline .
LIG1 expression levels predict treatment outcomes in:
Bladder Cancer: High expression correlates with reduced survival and chemotherapy resistance .
Ovarian Cancer: Elevated LIG1 levels are associated with platinum resistance .
PARP Inhibitors: LIG1 knockdown enhances the efficacy of PARP inhibitors in prostate cancer .
Immunotherapy: LIG1 inhibitors may improve responses to checkpoint inhibitors in bladder cancer .
DNA Ligase I (LIG1) is an ATP-dependent enzyme that plays a critical role in joining DNA strand breaks during DNA replication and repair processes, thus contributing significantly to genome integrity . It catalyzes the formation of phosphodiester bonds in the final step of DNA replication and certain DNA repair pathways. Recent studies have demonstrated that LIG1 not only functions in basic DNA metabolism but also appears to have roles in cancer progression, particularly in bladder cancer where it promotes malignant progression through enhancement of proliferation, invasion, and epithelial-mesenchymal transition (EMT) . Interestingly, contrary to earlier beliefs, some research has shown that LIG1 is not absolutely essential for mammalian cell viability in certain cell types, suggesting potential redundancy with other DNA ligases under specific cellular conditions .
Distinguishing between different DNA ligases (LIG1, LIG3, and LIG4) requires specific approaches:
Antibody specificity: Use validated antibodies targeting unique regions of each ligase. For example, the 67840-1-Ig LIG1 antibody is raised against the C-terminal 670-919 amino acid residues of human DNA ligase 1, providing specificity .
Molecular weight differentiation: LIG1 has a calculated molecular weight of 102 kDa but is observed at approximately 130 kDa on immunoblots due to post-translational modifications .
Adenylation assays: These assays can be performed to distinguish ligase activities. Cell extracts are incubated with α-32P-ATP, and the reaction mixture is resolved on SDS-PAGE gels to visualize adenylated ligase enzymes .
Gene targeting approaches: As demonstrated in experimental studies, targeted deletion of specific exons (such as exons 18-19 of the LIG1 gene) followed by confirmation via Southern and Northern blot analyses can validate the specificity of ligase-related effects .
Based on validated testing, LIG1 antibodies are most effectively used in the following applications:
The antibody has been successfully tested in multiple cell lines, including HepG2, HeLa, Jurkat, MOLT-4, K-562, and HSC-T6 cells, showing consistent reactivity across human, mouse, and rat samples . For optimal results, researchers should titrate the antibody concentration based on their specific experimental system.
Based on established methodologies, researchers should consider the following approach for LIG1 gene targeting experiments:
Design targeting vectors: Construct vectors containing homology blocks (approximately 2 kb DNA fragments) amplified from genomic DNA .
Select appropriate cell lines: Choose cell lines relevant to your research question. For example, CH12F3 mouse B cell line has been successfully used for LIG1 gene targeting .
Two-step targeting strategy: For complete knockout, perform sequential targeting of both alleles. First, create heterozygous cells (+/P) through puromycin selection, then proceed to the second round of targeting .
Validation methods:
Functional assays: Depending on the research question, design appropriate functional assays (e.g., cell viability, DNA repair capacity, or specific pathway analyses) to assess the consequences of LIG1 deficiency .
For robust Western blot experiments with LIG1 antibodies, researchers should include:
Positive controls: Cell lysates from cell lines known to express LIG1, such as HepG2, HeLa, Jurkat, MOLT-4, K-562, or HSC-T6 cells .
Negative controls:
Loading controls: β-actin or other housekeeping proteins to normalize protein loading .
Molecular weight markers: To confirm the expected 130 kDa band size (note the difference between calculated 102 kDa and observed 130 kDa molecular weight) .
Antibody titration: A range of antibody dilutions (e.g., 1:5000 to 1:50000) to determine optimal signal-to-noise ratio for your specific samples .
Several methodological approaches have been validated for studying LIG1 function:
Proliferation assays:
Migration and invasion assays:
Apoptosis analysis:
Drug sensitivity testing:
DNA repair capacity assessment:
Recent research has revealed significant correlations between LIG1 expression and both cancer prognosis and therapy response:
These findings suggest that LIG1 expression levels could serve as a biomarker for predicting treatment responses, particularly for immunotherapy approaches in cancer management.
The understanding of LIG1's essentiality has evolved significantly:
Historical perspective: LIG1 was previously reported to be essential for the viability of mouse embryonic stem cells (ESCs) .
Recent contradictory findings:
Cell type-specific requirements: The cellular lethality caused by LIG1 deficiency appears to be cell type-specific, with some cell lineages showing complete viability despite LIG1 absence .
Experimental validation: Gene targeting experiments involving deletion of exons 18-19 of the LIG1 gene, which causes frameshift mutations in all downstream exons, have confirmed the viability of certain LIG1-deficient cell types .
These findings suggest that the essentiality of LIG1 must be evaluated in a cell type-specific context, with potential compensation mechanisms existing in certain cellular lineages but not others.
Based on recent findings connecting LIG1 to immune regulation, researchers can employ these methodological approaches:
Immune infiltration analysis: Comprehensive analysis of tumor-infiltrating immune cells in relation to LIG1 expression levels, as this has been shown to impact prognosis in bladder cancer .
Mutational landscape assessment: Analysis of somatic mutations and copy number variations (CNV) between low and high LIG1 expression groups to identify potential mechanisms underlying differential immune responses .
Immunotherapy response prediction: Utilization of multiple cohorts (e.g., KIM, IMvigor210, Amato, GAO, Lauss, Riaz, and Ascierto) to validate correlations between LIG1 expression and response to various immunotherapies .
In vitro immune function assays: Following LIG1 knockdown or overexpression, researchers can assess:
Cytokine production profiles
Immune checkpoint molecule expression
Tumor cell recognition by immune effector cells
Drug sensitivity testing: Evaluation of response to immune checkpoint inhibitors and combination therapies in relation to LIG1 expression levels .
These approaches can help elucidate the mechanisms by which LIG1 influences immune cell recruitment and regulation within the tumor microenvironment.
Researchers commonly encounter these challenges when working with LIG1 antibodies:
Variable molecular weight detection:
Background signal in Western blots:
Cross-reactivity concerns:
Storage-related sensitivity loss:
Sample-dependent variability:
For successful co-immunoprecipitation (CoIP) experiments with LIG1 antibodies:
Pre-clearing step: Pre-clear lysates with protein A/G beads to reduce non-specific binding.
Antibody amount optimization: Titrate antibody amounts (typically starting with 2-5 μg per reaction) to determine optimal signal-to-noise ratio.
Incubation conditions: Perform antibody-lysate incubation at 4°C overnight with gentle rotation to maximize specific interactions while minimizing non-specific binding.
Washing stringency: Optimize wash buffer composition and washing steps to balance between maintaining specific interactions and reducing background.
Controls to include:
Detection method: Use highly sensitive detection methods for Western blot analysis of immunoprecipitated complexes, with appropriate dilution of detection antibodies .
When selecting a LIG1 antibody for research applications, consider:
Target epitope location:
Validation data comprehensiveness:
Antibody format and characteristics:
Application-specific performance:
Storage requirements and stability:
Recent advances in LIG1 research have significantly contributed to cancer biology understanding:
These findings point to LIG1 as a multifaceted contributor to cancer biology, with potential applications in prognostic assessment, treatment selection, and development of novel targeted therapies.
The discovery that LIG1's essentiality varies across cell types has several important implications:
These implications highlight the importance of context-specific evaluation of LIG1 function and the potential for leveraging cell type-specific dependencies in therapeutic development.
Several emerging technologies show promise for advancing LIG1 research:
CRISPR-based approaches:
CRISPR interference (CRISPRi) for precise temporal control of LIG1 expression
Base editing for introducing specific mutations to study structure-function relationships
CRISPR screens to identify synthetic lethal interactions with LIG1 deficiency
Single-cell technologies:
Single-cell RNA-seq to characterize cell type-specific consequences of LIG1 manipulation
Single-cell proteomics to assess protein-level changes in response to LIG1 perturbation
Live-cell imaging of fluorescently tagged LIG1 to study dynamics during DNA replication and repair
Structural biology advances:
Cryo-EM for high-resolution structural analysis of LIG1 complexes
AlphaFold and other AI-based structure prediction tools to model LIG1 interactions
Multi-omics integration:
Combined analysis of genomic, transcriptomic, and proteomic data to understand LIG1 regulatory networks
Pathway analysis to place LIG1 in the context of broader cellular processes
Organoid and patient-derived models:
Use of 3D organoid cultures to study LIG1 function in more physiologically relevant contexts
Patient-derived xenografts to evaluate LIG1-targeted therapies in personalized medicine approaches
These technologies offer opportunities to deepen our understanding of LIG1 biology and accelerate the translation of this knowledge into clinical applications.