The DCLRE1C antibody is a research reagent designed to detect the ARTEMIS protein, encoded by the DCLRE1C gene. This protein plays a critical role in V(D)J recombination (a process essential for generating diverse immune receptors) and DNA repair mechanisms via non-homologous end joining (NHEJ) . The antibody is widely used in immunological and genetic studies to assess ARTEMIS expression, activity, and mutations linked to immunodeficiencies.
Detects DCLRE1C in HeLa cells and human kidney tissue at dilutions of 1:500–1:1000 .
Used to study ARTEMIS expression in immunodeficiency models (e.g., SCID, Omenn syndrome) .
Identifies defective ARTEMIS protein in patients with V(D)J recombination defects (e.g., reduced T-cell receptor diversity) .
Mutations in DCLRE1C (e.g., c.194C>T, c.465-1G>C) cause SCID and Omenn syndrome by impairing V(D)J recombination .
The antibody aids in diagnosing these conditions by detecting truncated or absent ARTEMIS protein .
Dclre1c-knockout mice exhibit SCID-like phenotypes (reduced T/B cells, impaired lymphoid organs) and are used for human tumor xenograft studies .
The antibody facilitates validation of ARTEMIS deficiency in these models .
ARTEMIS resolves hairpin structures during V(D)J recombination and repairs ionizing radiation-induced DNA breaks .
Antibody-based assays confirmed its role in NHEJ and cell cycle regulation .
Protein Function:
DCLRE1C (ARTEMIS) is a nuclease with intrinsic 5′-3′ exonuclease activity on single-stranded DNA. After phosphorylation by and in complex with DNA-dependent protein kinase catalytic subunit, ARTEMIS acquires endonuclease activity on 5′ and 3′ overhangs, and hairpins. It is essential for opening hairpins during V(D)J recombination of immunoglobulin and T-cell receptor genes in lymphocyte development .
The significance of DCLRE1C in immunological research stems from its critical role in immune system development. Null mutations cause severe combined immunodeficiency (SCID) with a block in B- and T-cell development, while hypomorphic mutations can result in a spectrum of immunodeficiency phenotypes ranging from atypical SCID to antibody deficiency . This protein provides a crucial link between DNA repair mechanisms and immune system function.
Although the calculated molecular weight of full-length DCLRE1C (692 amino acids) is approximately 78 kDa, the observed molecular weight in Western blot analysis is typically around 60 kDa . This discrepancy between calculated and observed molecular weights is not uncommon for proteins and may be attributed to:
Protein folding affecting migration in SDS-PAGE
Post-translational modifications
Proteolytic processing of the full-length protein
When performing Western blot analysis, it's important to note that different isoforms may appear at different molecular weights, and truncated variants resulting from mutations might show altered migration patterns .
DCLRE1C has multiple transcript isoforms resulting from alternative splicing. The three main isoforms are:
| Isoform | Amino Acid Length | Functional Activity |
|---|---|---|
| a (canonical) | 692 | High functional activity |
| b | 577 | Limited functional activity |
| c | 572 | Limited functional activity |
Functional analysis demonstrated that only isoform a (the 692aa canonical sequence) induced high levels of GFP expression in recombination assays, similar to the murine Dclre1c transcript isoform a . Therefore, for most research applications, antibodies targeting isoform a would be most appropriate for studying the functional aspects of DCLRE1C.
When selecting antibodies, consider whether they target domains present in all isoforms of interest or are specific to particular isoforms.
Thorough validation of antibody specificity is essential for obtaining reliable results. Follow these methodological approaches:
Positive and negative controls:
Western blot validation:
Verify a single band at the expected molecular weight (~60 kDa)
For polyclonal antibodies, some minor bands may be acceptable but should be consistent
Multiple antibody approach:
Compare results using antibodies targeting different epitopes of DCLRE1C (N-terminal versus C-terminal)
Consistent results with different antibodies provide stronger validation
Peptide competition assay:
Pre-incubate antibody with the immunizing peptide
Signal should be significantly reduced when the antibody is blocked with its target peptide
Functional correlation:
Correlate protein detection with functional assays (V(D)J recombination activity, DNA repair capacity)
This is particularly important when studying mutations that may affect epitope recognition
Optimal sample preparation varies depending on the application:
Use recommended dilutions ranging from 1:500 to 1:2000 depending on the specific antibody
Include protease inhibitors in lysis buffers to prevent degradation
Consider preparing nuclear fractions as DCLRE1C is predominantly nuclear
Load 20-50 μg of total protein per lane
Fixation: 4% paraformaldehyde for cells or 10% neutral buffered formalin for tissues
Permeabilization: 0.1-0.5% Triton X-100 (10 minutes) to access nuclear antigens
Blocking: 5% BSA or normal serum to reduce background
Antibody incubation: Overnight at 4°C for optimal signal-to-noise ratio
Include nuclear counterstain (DAPI) to confirm nuclear localization
Thorough permeabilization is essential as DCLRE1C is a nuclear protein
Include appropriate isotype controls
Use Fc receptor blocking for immune cells to reduce non-specific binding
Detecting DCLRE1C in patient samples with hypomorphic mutations presents unique challenges as these mutations often result in reduced protein expression. Based on research findings, consider these approaches:
Antibody selection:
Choose antibodies targeting protein regions preserved in the specific mutations being studied
For C-terminal truncations, use antibodies targeting the N-terminal region
Enhanced detection methods:
Use high-sensitivity chemiluminescent substrates for Western blotting
Consider signal amplification systems for immunohistochemistry
Longer exposure times may be necessary, but control for increased background
Sample enrichment:
Immunoprecipitation to concentrate the protein before detection
Subcellular fractionation to enrich for nuclear proteins
Experimental controls:
Include samples from healthy donors processed identically
When available, include samples from patients with known null mutations as comparisons
Research has shown that ARTEMIS expression in fibroblasts from patients with the missense variant c.194C>T (p.T65I) was severely reduced compared to controls . Similar findings were observed in cells with compound heterozygous mutations. Western blot analyses of HEK293T cells transfected with plasmids encoding either missense or frameshift variants also yielded reduced expression of these ARTEMIS variants .
Several sophisticated experimental systems have been developed to study DCLRE1C function in V(D)J recombination:
Recombination reporter systems:
Retroviral constructs containing inverted GFP cassettes flanked by recombination signal sequences (RSS)
GFP expression serves as a functional read-out of recombination activity that can be quantified by flow cytometry
This system has been successfully used to assess the functional impact of DCLRE1C mutations
Cell cycle manipulation:
Complementation studies:
Dclre1c-/- cells can be transduced with wild-type or mutant DCLRE1C constructs
Functional recovery can be measured through recombination efficiency
This approach allows structure-function analysis of specific mutations
Analysis of recombination junctions:
Amplification and sequencing of immunoglobulin or T-cell receptor gene rearrangements
Examination of junctional diversity, palindromic nucleotide addition, and microhomology
Patients with DCLRE1C mutations show characteristic patterns, including increased palindromic nucleotides in the complementarity determining regions 3
DCLRE1C/ARTEMIS is involved in repairing a subset of DNA double-strand breaks through the NHEJ pathway. These methodological approaches can assess this function:
γH2AX resolution assays:
Colony survival assays:
Radiation sensitivity testing:
Comet assay:
Single-cell gel electrophoresis to measure DNA fragmentation
Comparison of tail moment before and after damage induction
These approaches can be applied to cell lines, patient-derived cells, or animal models to comprehensively characterize DCLRE1C's role in maintaining genomic stability.
When protein expression does not correlate with functional activity, consider these methodological approaches:
Isoform-specific analysis:
Post-translational modifications:
Protein-protein interactions:
DCLRE1C functions in complex with other proteins (DNA-PKcs, other NHEJ factors)
Co-immunoprecipitation can assess complex formation efficiency
Stability and turnover:
Some mutations affect protein stability rather than intrinsic activity
Pulse-chase experiments can determine protein half-life
Overexpression strategies:
Research has revealed important correlations between DCLRE1C expression patterns and clinical phenotypes:
Expression-phenotype spectrum:
Cellular manifestations:
B-cell lymphopenia is a consistent finding across phenotypes
T-cell counts may be normal despite functional deficits
Reduced naive T cells with increased terminally differentiated T cells indicate compensatory peripheral expansion
Restricted T-cell receptor repertoire reflects impaired V(D)J recombination
Laboratory parameters to monitor:
Geographic and founder mutations:
Working with patient samples presents different challenges compared to established cell lines:
Sample limitations:
Limited quantity and availability of patient material
Variability between patients, even with identical mutations
Need for appropriate controls (family members, age-matched healthy donors)
Cell type considerations:
Functional assessment:
Colony survival after irradiation provides a functional readout in patient cells
T-cell receptor repertoire analysis can assess the impact on V(D)J recombination in vivo
V(D)J recombination reporter assays may require transfer of patient-derived mutations to experimental systems
Standardization:
Process patient and control samples simultaneously
Establish clear cutoffs for normal versus abnormal results
Consider the effect of prior treatments (e.g., immunoglobulin replacement) on results
Ethical considerations:
Obtain appropriate informed consent for research use of samples
Consider genetic counseling implications when novel mutations are identified
Multiple bands or unexpected molecular weights in Western blots can occur for several reasons:
Alternative splicing:
The three major isoforms (a, b, c) have different molecular weights
Additional splice variants may exist in specific cell types
Truncated proteins from mutations:
Post-translational modifications:
Phosphorylation by DNA-PKcs changes the migration pattern
Other modifications may occur in response to DNA damage or cell cycle progression
Degradation products:
DCLRE1C may be subject to regulated proteolysis
Insufficient protease inhibition during sample preparation can cause artifactual bands
Antibody cross-reactivity:
Some antibodies may recognize similar epitopes in related proteins
Validate with knockout controls or peptide competition assays
Technical issues:
Insufficient blocking leading to non-specific binding
Excessive antibody concentration
Over-development of the blot
Working with primary immune cells presents specific challenges:
Low expression levels:
DCLRE1C expression may be lower in primary cells compared to cell lines
Signal amplification methods may be necessary
Longer exposure times for Western blots or increased antibody concentration for immunostaining
Cell heterogeneity:
Mixed cell populations in blood or tissue samples
Consider cell sorting or co-staining with lineage markers
Single-cell approaches may reveal population heterogeneity
Activation state effects:
Expression and localization may change with cellular activation
Document activation status of cells (resting vs. stimulated)
Compare equivalent activation states between samples
Limited cell numbers:
Optimize protocols for small sample sizes
Consider nested PCR approaches for transcript analysis
Use micro-Western techniques for protein detection
Background issues in flow cytometry:
Autofluorescence of primary cells, especially after fixation
Include proper controls (unstained, isotype, FMO)
Use Fc receptor blocking reagents to reduce non-specific binding