DCLRE1C Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Purpose

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.

Applications in Research

Western Blotting:

  • 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) .

ELISA:

  • Validates antibody specificity for DCLRE1C in serum or lysate samples .

Immunological Studies:

  • Identifies defective ARTEMIS protein in patients with V(D)J recombination defects (e.g., reduced T-cell receptor diversity) .

  • Assesses NHEJ efficiency in DNA repair assays .

Immunodeficiency Diagnostics

  • 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 .

Mouse Models

  • 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 .

DNA Repair Insights

  • 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 .

Structure and Function of ARTEMIS

  • Gene Location: Chromosome 10 (NC_000010.11) .

  • Protein Function:

    • 5'–3' exonuclease activity on single-stranded DNA .

    • Endonuclease activity on hairpins and overhangs (requires PRKDC/DNA-PKcs) .

Clinical Relevance

  • Diseases: SCID (T-B-NK+), Omenn syndrome, Hyper IgM syndrome .

  • Therapeutic Monitoring: The antibody supports gene therapy and bone marrow transplant evaluations by quantifying ARTEMIS expression .

Product Specs

Buffer
PBS with 0.1% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze/thaw cycles.
Lead Time
Typically, we can ship your order within 1-3 business days of receiving it. Delivery times may vary depending on the purchase method and location. Please consult your local distributor for specific delivery time information.
Synonyms
A SCID antibody; A SCID protein antibody; Artemis protein antibody; ASCID antibody; DCLRE1C antibody; DCLRE1C DNA cross link repair 1C antibody; DCLRE1C protein antibody; DCLREC1C antibody; DCR1C_HUMAN antibody; DNA cross link repair 1C antibody; DNA cross link repair 1C protein antibody; DNA cross-link repair 1C protein antibody; FLJ11360 antibody; FLJ36438 antibody; hSNM1C antibody; OTTHUMP00000045150 antibody; Protein A-SCID antibody; Protein ARTEMIS antibody; PSO2 homolog antibody; RS SCID antibody; SCIDA antibody; Severe combined immunodeficiency type a antibody; SNM1 homolog C antibody; SNM1 like protein antibody; SNM1-like protein antibody; SNM1C antibody
Target Names
DCLRE1C
Uniprot No.

Target Background

Function
Artemis is a nuclease that plays a crucial role in DNA non-homologous end joining (NHEJ), a major DNA repair pathway. NHEJ is essential for the repair of double-strand breaks (DSBs) and V(D)J recombination, the process that generates the diverse repertoire of antigen-binding domains in immunoglobulins and T-cell receptors. During V(D)J recombination, the RAG endonuclease complex generates specific DSBs, resulting in two distinct DNA end structures: hairpin sealed coding ends and phosphorylated blunt signal ends. Artemis, in conjunction with other NHEJ factors, repairs these ends, forming coding and signal joints respectively. In isolation, Artemis exhibits single-strand specific 5'-3' exonuclease activity. However, when complexed with DNA-PKcs, it acquires endonucleolytic activity on 5' and 3' hairpins and overhangs. This activity is crucial for the resolution of closed hairpins before coding joint formation. Artemis is also involved in the repair of complex DSBs induced by ionizing radiation, which require extensive end processing prior to religation by NHEJ.
Gene References Into Functions
  1. Research suggests that the activation of Artemis nuclease by the XRCC4-DNA ligase IV complex and the efficiency of blunt-end ligation are influenced by the structural configurations at the DNA ends. (XRCC4 = X-ray repair cross complementing 4) PMID: 28696258
  2. An N-terminal fragment encompassing the catalytic domain interacts with both itself and a C-terminal fragment. Amino acid exchanges N456A+S457A+E458Q in the C-terminus of full-length SCIDA resulted in the exposure of the N-terminus and increased SCIDA activity in cellular V(D)J recombination assays. PMID: 28082683
  3. Data demonstrate that DCLRE1C mutations can cause a phenotype presenting as antibody deficiency alone. PMID: 26476407
  4. Mutations in DCLRE1C and NCF1 have been identified through whole-genome sequencing in unrelated patients with primary immunodeficiency. PMID: 25981738
  5. The nature and location of DCLRE1C mutations correlate with the clinical phenotype of severe combined immunodeficiency. PMID: 25917813
  6. Studies have identified Artemis as a PTIP-binding protein. PMID: 25512557
  7. The 5'-exonuclease activity is intrinsic to ARTEMIS, indicating its relevance to the role of ARTEMIS in nonhomologous DNA end joining. PMID: 24500713
  8. DNA ligase IV and Artemis collaborate to promote nonhomologous end-joining. PMID: 23967291
  9. Two siblings have been described with combined immunodeficiency (CID) and immunodysregulation caused by compound heterozygous Artemis mutations. PMID: 24230999
  10. Artemis levels have a significant impact on radiation toxicity in human cells. PMID: 22713703
  11. Research suggests that Artemis functions as a molecular switch that converts stalled replication forks containing single-stranded gap DNA lesions into double-strand breaks, thereby activating the ATM signaling pathway. PMID: 23465063
  12. Structural insights into the DNA ligase IV-Artemis interaction in nonhomologous end-joining have been revealed. PMID: 23219551
  13. These findings indicate that Artemis acts as a positive regulator of AMPK signaling by stabilizing the LKB1-AMPK complex. PMID: 23044421
  14. A study identified a novel SCID mutation in a consanguineous Israeli Arab family. Sequencing revealed an 8 bp insertion in exon 14 (1306ins8) of DCLRE1C in all affected patients. This insertion causes an alteration in amino acid 330 of the protein from cysteine to a stop codon (p.C330X). PMID: 22527898
  15. Results show that Artemis, but not ATM, is dispensable for homologous recombination of radiation-induced double-strand breaks during S-phase. PMID: 22730303
  16. The regulation of p27 by Artemis and DDB2 is essential for cell cycle progression in normally proliferating cells. PMID: 22134138
  17. Point mutations in Artemis that disrupt its interaction with Ligase IV or DNA-PKcs reduce V(D)J recombination. PMID: 22529269
  18. The dominant negative mutant Artemis fragment (D37N-413aa) enhanced tumor cell radiosensitivity by inhibiting the activity of endogenous Artemis and DNA repair. PMID: 21641068
  19. Antisense oligonucleotide (AON) targeting the intronic mutation restored wild-type Artemis transcript levels and non-homologous end-joining pathway activity in patient fibroblasts. PMID: 21390052
  20. Analysis of differences in sensitivity to DNA-damaging agents between XRCC4- and Artemis-deficient human cells has been conducted. PMID: 21785230
  21. Artemis is essential for the repair of DNA double-strand breaks arising endogenously or following oxidative stress. PMID: 21596788
  22. Restoration of chemo/radioresistance by wild-type, but not D165N Artemis, suggests that the lack of endonucleolytic trimming of DNA ends is the primary cause of sensitivity to double-strand cleaving agents in Artemis-deficient cells. PMID: 21531702
  23. Studies indicate that codon-based models of gene evolution show statistical support for the recurrent positive selection of five NHEJ genes during primate evolution: XRCC4, NBS1, Artemis, POLlambda, and CtIP. PMID: 20975951
  24. Functional analyses on patient fibroblasts demonstrated that the corresponding alleles carry null mutations of the DCLRE1C gene. PMID: 19953608
  25. Artemis plays a role in the 3'-processing reaction and protection of the ends of viral DNA (HIV-1) after reverse transcription. It is involved in multiple steps, including integration and pre-integration, during retroviral replication. PMID: 20003485
  26. DNA-PKcs regulates Artemis through both phosphorylation and complex formation, enabling enzymatic activities crucial for the hairpin-opening step of V(D)J recombination and the 5' and 3' overhang processing in nonhomologous DNA end joining. PMID: 11955432
  27. A nonsense founder mutation identified in exon 8 of Artemis results in the truncation of the deduced protein product. This finding suggests that the SNM1-like gene (Artemis) is responsible for SCID in Athabascan-speaking Native Americans. PMID: 12055248
  28. Deletions and missense mutations in the Artemis gene can cause radiosensitive-SCID, characterized by defective coding joint formation and an early and complete B-cell differentiation block. PMID: 12406895
  29. Artemis plays a role in T and B lymphocyte immunodeficiency and in predisposition to lymphoma through the NHEJ pathway of DNA repair. PMID: 12569164
  30. The genomic exon 3 deletion is unique to Japan and is considered a founder haplotype. PMID: 12592555
  31. The properties of Artemis proteins are integrated into the processes of V(D)J recombination and non-homologous end-joining. PMID: 14628082
  32. Artemis utilizes one or two Zn(II) ions to exert its catalytic activity, similar to bacterial class B beta-Lact enzymes that hydrolyze beta-lactam compounds. PMID: 14744996
  33. The hairpin-opening activity of ARTEMIS, and/or its overhang endonucleolytic activity, are necessary for V(D)J recombination; however, its exonuclease activity alone is insufficient for this process. PMID: 15071507
  34. Data show that Artemis interacts with cell cycle checkpoint proteins and is a phosphorylation target of the checkpoint kinases ATM or ATR following exposure of cells to IR or UV irradiation, respectively. PMID: 15456891
  35. Artemis is an effector of DNA repair that can be phosphorylated by ataxia-telangiectasia-mutated kinase (ATM) and potentially by DNA-dependent protein kinase catalytic subunit and ATM-and Rad3-related kinase, depending on the type of DNA damage. PMID: 15468306
  36. ATM, Artemis, and proteins localized to gamma-H2AX foci play roles in double-strand break rejoining. PMID: 15574327
  37. Research findings indicate that Artemis is positioned at the signaling crossroads downstream of DNA-PKcs and ATM in IR-induced DSB repair. PMID: 15723659
  38. A report describes the first patient with clinical and immunological features of OS caused by hypomorphic ARTEMIS mutations. Sequencing of the ARTEMIS gene revealed a compound heterozygosity in this nonhomologous end-joining (NHEJ) factor. PMID: 15731174
  39. The Artemis:DNA-PKcs nuclease might be important in removing secondary structures that hinder the processing of DNA ends during nonhomologous DNA end joining. PMID: 15936993
  40. The uncharacterized C-terminal domain of Artemis has important regulatory roles; results lead to a model for how DNA-PKcs activates Artemis through phosphorylation. PMID: 16093244
  41. Six DNA-PK phosphorylation sites on Artemis have been characterized. Phosphorylation of these sites depends on the association with DNA-PK catalytic subunit and is induced by double-stranded DNA damage. PMID: 16600297
  42. Ku-mediated assembly of DNA-PK on DNA ends leads to the dissociation of the DNA-PKcs.Artemis complex. PMID: 16857680
  43. DNA-PK autophosphorylation regulates Artemis access to DNA ends, providing insight into the mechanism of Artemis-mediated DNA end processing. PMID: 16874298
  44. Artemis efficiently trims long 3'-phosphoglycate-terminated overhangs induced in DNA by radiation and other radical-based toxins. PMID: 17121861
  45. The phenotype of cells derived from SCID patients with different mutations in the Artemis gene has been analyzed. PMID: 17169382
  46. ATM regulates G(2)/M checkpoint recovery through inhibitory phosphorylations of Artemis that occur shortly after DNA damage. This sets a molecular switch that, hours later upon completion of DNA repair, allows the activation of the Cdk1-cyclin B complex. PMID: 17242184
  47. There is some sequence-dependent variation in the efficiency and position of hairpin opening by Artemis:DNA-PKcs, providing more clarity on the extent to which the hairpin opening position contributes to junctional diversity in V(D)J recombination. PMID: 17932067
  48. The Artemis C-terminus is essential for V(D)J recombination at the normal Artemis expression level. PMID: 18034425
  49. H254 plays a key role in Artemis function, as it is critical for its full activity in vitro. PMID: 19022407
  50. Results link Artemis to the predominant nonhomologous end-joining pathway during immunoglobulin class switch recombination. PMID: 19075292

Show More

Hide All

Database Links

HGNC: 17642

OMIM: 602450

KEGG: hsa:64421

STRING: 9606.ENSP00000367527

UniGene: Hs.655932

Involvement In Disease
Severe combined immunodeficiency autosomal recessive T-cell-negative/B-cell-negative/NK-cell-positive with sensitivity to ionizing radiation (RSSCID); Severe combined immunodeficiency Athabaskan type (SCIDA); Omenn syndrome (OS)
Protein Families
DNA repair metallo-beta-lactamase (DRMBL) family
Subcellular Location
Nucleus.
Tissue Specificity
Ubiquitously expressed, with highest levels in the kidney, lung, pancreas and placenta (at the mRNA level). Expression is not increased in thymus or bone marrow, sites of V(D)J recombination.

Q&A

What is DCLRE1C and why is it significant in immunological research?

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.

What molecular weight should I expect for DCLRE1C in Western blot analyses?

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 .

What are the common isoforms of DCLRE1C and which should I target with antibodies?

DCLRE1C has multiple transcript isoforms resulting from alternative splicing. The three main isoforms are:

IsoformAmino Acid LengthFunctional Activity
a (canonical)692High functional activity
b577Limited functional activity
c572Limited 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.

How should I validate the specificity of a DCLRE1C antibody for my research?

Thorough validation of antibody specificity is essential for obtaining reliable results. Follow these methodological approaches:

  • Positive and negative controls:

    • Use known positive samples such as HeLa cells or human kidney tissue, which have been demonstrated to express DCLRE1C

    • Include DCLRE1C knockout or knockdown samples as negative controls

    • Mouse thymus has been identified as a positive sample for DCLRE1C detection

  • 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

What are optimal sample preparation methods for detecting DCLRE1C in different experimental settings?

Optimal sample preparation varies depending on the application:

For Western Blotting:

  • 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

For Immunohistochemistry/Immunofluorescence:

  • 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

For Flow Cytometry:

  • 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

How can I optimize detection of DCLRE1C in patient samples with hypomorphic mutations?

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 .

What experimental approaches can be used to study DCLRE1C function in relation to V(D)J recombination?

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:

    • V-abl kinase transformed murine Dclre1c-/- pro-B cells can be blocked in G0/G1 cell cycle phases using imatinib (3μM)

    • This allows the Rag1/Rag2 complex to initiate recombination with higher efficiency

  • 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

How can I assess DCLRE1C's role in DNA repair beyond V(D)J recombination?

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:

    • Measure γH2AX levels at 1h, 8h, 24h, and 36h after ionizing radiation (10Gy)

    • DNA repair efficiencies can be calculated based on mean fluorescent intensities at 36h after irradiation

    • Cells with defective DCLRE1C show impaired resolution of γH2AX foci

  • Colony survival assays:

    • Assess cell survival after exposure to ionizing radiation

    • Patients with DCLRE1C mutations show decreased colony survival after irradiation

  • Radiation sensitivity testing:

    • ARTEMIS-deficient cells exhibit increased sensitivity to double-strand breaks induced by ionizing radiation or chemical agents like bleomycin and neocarzinostatin

    • Dose-response curves can quantify this sensitivity

  • 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.

What approaches help resolve discrepancies between DCLRE1C protein expression and functional activity?

When protein expression does not correlate with functional activity, consider these methodological approaches:

  • Isoform-specific analysis:

    • Only isoform a (692aa) shows high functional activity, while isoforms b and c have limited activity

    • Antibodies detecting all isoforms might not reflect functional capacity

  • Post-translational modifications:

    • Phosphorylation by DNA-PKcs is required for DCLRE1C activation

    • Use phospho-specific antibodies (e.g., targeting pSer516) to assess activation status

  • 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 shown that "Overexpression of hypomorphic mutants may improve the functional defect"

    • Quantitative analysis can determine how much increased expression is needed to restore function

How does DCLRE1C expression correlate with clinical phenotypes in immunodeficiency?

Research has revealed important correlations between DCLRE1C expression patterns and clinical phenotypes:

  • Expression-phenotype spectrum:

    • Null mutations causing complete absence of functional protein typically result in T-B-NK+ SCID

    • Hypomorphic mutations allowing residual protein expression can cause milder phenotypes ranging from atypical SCID to antibody deficiency

  • 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:

    • Very low B-cell numbers and serum IgA levels are characteristic findings

    • T-cell proliferative responses to stimulation are typically impaired

    • Western blot analysis can demonstrate reduced ARTEMIS expression in patient fibroblasts

  • Geographic and founder mutations:

    • The c.194C>T (p.T65I) mutation has been identified in multiple patients from the same geographic region

    • Such founder mutations may be associated with specific phenotypic presentations

What technical considerations are important when analyzing DCLRE1C in patient samples versus cell lines?

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:

    • Fibroblasts have been successfully used for Western blot analysis of DCLRE1C expression

    • Peripheral blood lymphocytes can be analyzed for naive T-cell counts and proliferative responses

    • Immortalized patient cell lines may show altered expression patterns compared to primary cells

  • 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

Why might I observe multiple bands or unexpected molecular weights when using DCLRE1C antibodies?

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:

    • Frameshift or nonsense mutations produce truncated proteins

    • For example, the frameshift variant c.1669_1670insA (p.T577Nfs*21) produces a truncated protein visible as an additional band on Western blot

  • 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

What are common pitfalls when analyzing DCLRE1C in primary immune cells?

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

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 2025 TheBiotek. All Rights Reserved.