HELLS Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
PBS with 0.1% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze/thaw cycles.
Lead Time
We typically dispatch products within 1-3 business days after receiving your order. Delivery times may vary depending on the purchasing method or location. Please consult your local distributors for specific delivery time information.
Synonyms
FLJ10339 antibody; Helicase lymphoid specific antibody; Hells antibody; HELLS_HUMAN antibody; LSH antibody; Lymphoid specific helicase antibody; Lymphoid-specific helicase antibody; Nbla10143 antibody; PASG antibody; Proliferation associated SNF2 like protein antibody; Proliferation-associated SNF2-like protein antibody; SWI/SNF2 related matrix associated actin dependent regulator of chromatin subfamily A member 6 antibody; SWI/SNF2-related matrix-associated actin-dependent regulator of chromatin subfamily A member 6 antibody
Target Names
HELLS
Uniprot No.

Target Background

Function
HELLS plays a critical role in normal development and survival. It is involved in regulating the expansion or survival of lymphoid cells. HELLS is required for de novo or maintenance DNA methylation. It may control silencing of the imprinted CDKN1C gene through DNA methylation. Additionally, HELLS may play a role in the formation and organization of heterochromatin, suggesting a functional role in regulating transcription and mitosis.
Gene References Into Functions
  1. HELLS and CDCA7 form a bipartite nucleosome remodeling complex. Immunodeficiency-centromeric instability-facial anomalies syndrome is characterized by a defective HELLS and CDCA7 bipartite nucleosome remodeling complex. PMID: 29339483
  2. This research elucidates the molecular basis of the c-Myc/EGLN1-mediated induction of LSH expression that inhibits ferroptosis. PMID: 28900510
  3. LSH is likely one of the mechanisms responsible for genome instability underlying 5-hydroxymethylcytosine loss in cancer. PMID: 29109788
  4. Elevated mRNA levels of HELLS are an independent predictor of poor prognosis in Renal Cell Carcinoma patients. PMID: 28069330
  5. Mutations in HELLS cause immunodeficiency-centromeric instability-facial anomalies syndrome type 4. PMID: 26216346
  6. Mutations of the HELLS gene are associated with stomach and colorectal cancers. PMID: 25351940
  7. Upregulation of HELLS and UHRF1 is essential for the tumor phenotype. These epigenetic regulators are also important for the regulation of SYK. PMID: 25338120
  8. Similar to E2F3, HELLS is overexpressed in human tumors, including prostate cancer, indicating that either factor may contribute to the malignant progression of tumors. PMID: 22157815
  9. E2F1 plays a crucial role in transcriptional control of the human Lsh gene, and the decrease in Lsh expression in senescent cells is related to the repression of E2F1. PMID: 21453717
  10. Data provide strong evidence that CEP55 and HELLS may be used in conjunction with FOXM1 as a biomarker set for early cancer detection and indicators of malignant conversion and progression. PMID: 20400365
  11. LSH serves as a recruiting factor for DNA methyltransferases and histone deacetylases to establish transcriptionally repressive chromatin, which is perhaps further stabilized by DNA methylation at targeted loci. PMID: 17967891
  12. These findings suggest differences in the cellular consequences of hypomethylation mediated by PASG during development compared to that in somatic cells. PMID: 18948754
  13. Lsh overexpression delays cell senescence by silencing p16(INK4a) in human fibroblasts. PMID: 19561196

Show More

Hide All

Database Links

HGNC: 4861

OMIM: 603946

KEGG: hsa:3070

STRING: 9606.ENSP00000239027

UniGene: Hs.463677

Involvement In Disease
Immunodeficiency-centromeric instability-facial anomalies syndrome 4 (ICF4)
Protein Families
SNF2/RAD54 helicase family
Subcellular Location
Nucleus.
Tissue Specificity
Highly expressed in proliferative tissues such as adult thymus and testis, and expressed at lower levels in uterus, small intestine, colon, and peripheral blood mononuclear cells. Also expressed in neoplastic cell lines including those derived from myeloi

Q&A

What is HELLS/LSH and how does it function in cellular processes?

HELLS (Helicase, Lymphoid Specific), also known as LSH (Lymphoid-specific helicase), is a SNF2-like chromatin remodelling protein primarily involved in DNA methylation processes. It functions as a critical regulator of chromatin structure and accessibility that plays essential roles in:

  • De novo DNA methylation and methylation maintenance, particularly at repetitive sequences

  • Heterochromatin formation and organization

  • Transcriptional regulation through chromatin remodeling

  • DNA double-strand break repair, particularly in heterochromatic regions

  • B-cell development and germinal center reactions

  • Class switch recombination in B lymphocytes

Loss-of-function mutations in the HELLS gene in humans cause ICF4 syndrome (Immunodeficiency, Centromeric instability, Facial anomalies), characterized by hypogammaglobulinemia and severe recurrent infections .

What applications are HELLS antibodies most commonly validated for in research?

HELLS antibodies have been validated for multiple experimental applications with varying levels of reliability depending on the specific antibody and manufacturer. Based on the available data:

ApplicationValidation StatusCommon Uses
Western Blotting (WB)Widely validatedDetection of endogenous HELLS (97 kDa)
Immunohistochemistry (IHC)Validated by some manufacturersTissue expression patterns
Immunocytochemistry/Immunofluorescence (ICC-IF)Validated in specific antibodiesSubcellular localization
Chromatin Immunoprecipitation (ChIP)Validated for specific antibodiesIdentifying HELLS-bound genomic regions
ELISACommonly validatedQuantitative detection

When selecting an antibody, researchers should verify that the specific application they intend to use has been validated by the manufacturer. For instance, Cell Signaling Technology's HELLS Antibody (#7998) is primarily validated for Western Blotting applications with 1:1000 recommended dilution .

How should researchers design experiments to study HELLS function in B-cell development?

Based on recent literature, the most effective approach to study HELLS function in B-cell development involves conditional knockout mouse models, as constitutive deletion of HELLS is lethal. The experimental design should include:

  • Mouse Model Selection:

    • B-cell-specific conditional knockout models (Mb1-Cre Hells or CD21-Cre Hells) enable study of HELLS function at different B-cell developmental stages

    • Mb1-Cre Hells targets early B-cell development

    • CD21-Cre Hells targets mature naive B cells

  • Immunological Assessments:

    • Baseline immunoglobulin measurements by ELISA (IgM, IgG1, IgG2b, IgG3, IgA)

    • T-dependent antigen challenges using NP-CGG adsorbed on alum

    • Flow cytometry to analyze:

      • Germinal center B cells (B220+GL7+CD95+)

      • Memory B cell populations (B220+IgM-IgD-IgG1+CD38+GL7-)

      • Plasma cells (CD138+TACI+IgG1+)

  • Molecular Analysis:

    • DNA methylation profiling

    • V(D)J recombination assessment

    • Class-switch recombination analysis

    • Single-cell sequencing of rearranged immunoglobulin genes

Recent studies have demonstrated that HELLS-deficient mice show impaired germinal center reactions and premature decay of germinal center B cells, affecting long-term humoral immunity .

What are the optimal conditions for using HELLS antibodies in Western blotting applications?

For optimal Western blotting results with HELLS antibodies, researchers should consider the following protocol recommendations:

  • Sample Preparation:

    • Extract nuclear proteins (HELLS is a nuclear protein)

    • Use phosphatase inhibitors in lysis buffer as HELLS can be phosphorylated

    • Include protease inhibitors to prevent degradation

  • Electrophoresis Conditions:

    • Expect HELLS to migrate at approximately 97 kDa

    • Use 8-10% SDS-PAGE gels for optimal separation

  • Transfer and Detection:

    • Standard wet or semi-dry transfer to PVDF or nitrocellulose membranes

    • Blocking: 5% non-fat milk or BSA in TBST (1 hour at room temperature)

    • Primary antibody: 1:1000 dilution (as recommended for Cell Signaling Technology #7998)

    • Incubation: Overnight at 4°C

    • Secondary antibody: Anti-rabbit HRP conjugated (1:5000-1:10000)

    • Detection: Enhanced chemiluminescence (ECL)

  • Controls:

    • Positive control: HELLS-expressing cell lines (lymphoid cells recommended)

    • Negative control: HELLS-knockout or knockdown cells

    • Loading control: Nuclear proteins such as histone H3 or lamin B

  • Troubleshooting:

    • If multiple bands appear, optimize antibody concentration

    • For weak signals, extend exposure time or increase protein loading

    • Consider antigen retrieval methods if signal is weak despite confirmed expression

How do researchers accurately assess HELLS-dependent DNA methylation changes in experimental systems?

To accurately assess HELLS-dependent DNA methylation changes, researchers should employ a multi-faceted approach:

  • Genome-wide DNA Methylation Analysis:

    • Whole-genome bisulfite sequencing (WGBS) to map 5-methylcytosine at single-nucleotide resolution

    • Reduced representation bisulfite sequencing (RRBS) for cost-effective profiling

    • Methylated DNA immunoprecipitation sequencing (MeDIP-seq)

  • Locus-specific Analysis:

    • Bisulfite PCR followed by sequencing for candidate regions

    • Pyrosequencing for quantitative assessment of CpG methylation

    • Methylation-sensitive restriction enzyme analysis

  • Experimental Design Considerations:

    • Include appropriate controls (wild-type vs. HELLS-deficient)

    • Time-course experiments to distinguish between de novo methylation and maintenance effects

    • Cell type-specific analysis as HELLS effects may vary between tissues

  • Data Analysis:

    • Focus on repetitive DNA sequences (major and minor satellite sequences) where HELLS shows strong effects

    • Analyze both repeat regions and non-repeat sequences (single-copy genes)

    • Compare with gene expression data to correlate methylation changes with transcriptional outcomes

Research has shown that HELLS-deficient germinal center B cells undergo dramatic DNA hypomethylation and massive de-repression of evolutionary recent retrotransposons , suggesting that HELLS plays a critical role in DNA methylation maintenance during rapid proliferation.

What role does HELLS play in DNA damage repair, and how can this be experimentally investigated?

HELLS has been implicated in DNA double-strand break (DSB) repair, particularly in heterochromatic regions during G2 phase. To investigate this function:

  • Experimental Systems:

    • HELLS knockdown/knockout cell lines

    • Reconstitution with wild-type or ATP-binding site mutant HELLS (K254R)

    • Induction of DNA damage using ionizing radiation (IR) or site-specific endonucleases

  • Assays for DNA Repair Function:

    • DR-GFP assay to measure homologous recombination efficiency

    • Immunofluorescence monitoring of repair proteins (γH2AX, RAD51, CtIP)

    • Comet assay to assess DSB resolution kinetics

    • ChIP-sequencing to map HELLS localization at break sites

  • Mechanistic Investigations:

    • Co-immunoprecipitation to detect HELLS interactions with repair factors like CtIP

    • In vitro binding assays with recombinant proteins

    • ATP hydrolysis assays to assess chromatin remodeling activity

    • Chromatin accessibility assays (ATAC-seq) before and after damage

Research has shown that HELLS facilitates homologous recombination at two-ended breaks and contributes to repair within heterochromatic regions during G2 phase . The mechanism involves direct interaction with CtIP, promoting its accumulation at IR-induced breaks and subsequent end-resection. The ATPase activity of HELLS appears essential for this function, as the K254R mutant fails to rescue CtIP foci formation despite retaining CtIP binding capacity .

How does HELLS dysfunction contribute to ICF4 syndrome pathophysiology?

ICF4 syndrome is a rare inherited immunodeficiency resulting from loss-of-function mutations in the HELLS gene. The pathophysiological mechanisms include:

  • B-cell Developmental Defects:

    • Studies using conditional knockout mouse models have revealed that HELLS deficiency results in abnormal B lymphocyte development

    • Impaired germinal center formation and premature collapse of germinal center reactions

    • Reduced high-affinity memory B cell generation

    • Deficient long-term humoral immunity

  • Class Switch Recombination (CSR) Impairment:

    • HELLS is crucial for efficient immunoglobulin class switching

    • Deficiency leads to diminished IgG production

    • The defect appears not in the initiation of DNA double-stranded breaks but in the joining process during CSR

  • Hypogammaglobulinemia Mechanism:

    • HELLS-deficient mice show significant reduction in IgG1, IgG2b, and IgG3 titers

    • Primary IgG1 responses are initially normal but decline prematurely

    • Secondary (memory) IgG1 responses are severely impaired

    • This pattern explains the recurrent infections in ICF4 patients

  • DNA Methylation Abnormalities:

    • Hypomethylation of specific genomic regions, particularly repetitive elements

    • Instability of pericentromeric heterochromatin

    • Derepression of retrotransposons that may further destabilize the genome

This comprehensive understanding of how HELLS dysfunction impacts B-cell biology provides insights into potential therapeutic approaches for ICF4 patients, potentially focusing on restoring proper immune function or addressing the consequences of aberrant DNA methylation .

What experimental challenges arise when investigating HELLS in primary human samples from ICF4 patients?

Investigating HELLS in primary human samples from ICF4 patients presents several unique experimental challenges:

  • Sample Availability and Quality:

    • ICF4 is an extremely rare syndrome, limiting patient sample availability

    • Patients often have severe immunodeficiency, resulting in limited immune cell numbers

    • Samples may come from patients on various treatments, introducing confounding variables

  • Technical Considerations:

    • HELLS function assessment requires fresh cells for certain assays (e.g., class switch recombination)

    • DNA methylation patterns can be affected by sample processing and storage conditions

    • Patient cells may have compensatory mechanisms that mask direct HELLS effects

  • Analytical Approaches:

    • Comparison to appropriate controls is challenging due to patient genetic background variation

    • Need for single-cell approaches to address cellular heterogeneity

    • Integration of multiple omics datasets (genomics, epigenomics, transcriptomics) for comprehensive analysis

  • Methodological Solutions:

    • Establish patient-derived lymphoblastoid cell lines for renewable experimental material

    • Generate induced pluripotent stem cells (iPSCs) from patient samples for differentiation studies

    • Implement CRISPR/Cas9 gene editing to create isogenic control lines

    • Use of conditional HELLS knockout mouse models alongside patient samples for validation

  • Ethical Considerations:

    • Working with samples from vulnerable patient populations requires stringent ethical oversight

    • Limited sample availability necessitates maximizing data generation from each sample

    • Importance of returning clinically relevant findings to benefit patients when possible

How should researchers validate HELLS antibodies for specificity and sensitivity in their experimental systems?

Comprehensive validation of HELLS antibodies is crucial for experimental reliability. Researchers should implement the following validation strategy:

  • Knockout/Knockdown Controls:

    • Test antibodies on HELLS knockout or knockdown samples alongside wild-type controls

    • Use siRNA, shRNA, or CRISPR-Cas9 to generate HELLS-depleted controls

    • Compare multiple antibody clones on these control samples

  • Application-Specific Validation:

    • For Western blotting: Verify single band at expected molecular weight (97 kDa)

    • For immunofluorescence: Confirm nuclear localization pattern

    • For ChIP applications: Validate enrichment at known HELLS target loci

    • For immunohistochemistry: Compare with mRNA expression patterns

  • Cross-reactivity Assessment:

    • Test on tissues/cells from different species if cross-reactivity is claimed

    • Evaluate potential cross-reactivity with closely related helicases

    • Perform peptide competition assays to confirm epitope specificity

  • Documentation of Validation Data:

    • Record all validation experiments with appropriate controls

    • Document antibody catalog number, lot number, and experimental conditions

    • Share validation data with colleagues to establish consensus on antibody reliability

  • Common Pitfalls to Avoid:

    • Relying solely on manufacturer's validation data

    • Using a single application to validate for multiple applications

    • Failing to include proper positive and negative controls

This validation approach is particularly important given the variability in commercial antibodies and the challenges researchers face with antibody reproducibility .

What are the current limitations of HELLS antibodies in research applications and how can they be addressed?

Current HELLS antibodies present several limitations that researchers should be aware of and address:

  • Variability Between Lots and Manufacturers:

    • Different antibodies show variable specificity and performance

    • Limited standardization between manufacturers

    • Solution: Validate each new lot with positive and negative controls; maintain records of lot numbers used in experiments

  • Application Restrictions:

    • Many antibodies are only validated for specific applications (e.g., Western blot only)

    • Performance in complex tissues may differ from cell lines

    • Solution: Perform comprehensive validation for each intended application; use multiple antibodies targeting different epitopes

  • Species Cross-reactivity Limitations:

    • Not all antibodies work across multiple species

    • Claimed cross-reactivity may not be thoroughly validated

    • Solution: Validate antibodies specifically for each species of interest; consider species-specific antibodies for critical experiments

  • Technical Challenges:

    • High background in some applications (particularly IHC and IF)

    • Potential cross-reactivity with related helicases

    • Epitope masking in certain experimental conditions

    • Solution: Optimize blocking conditions; use monoclonal antibodies for higher specificity; test different epitope retrieval methods

  • Cost and Availability Issues:

    • High cost limits extensive validation and replicate experiments ($400-650 per vial)

    • Limited availability of certain specialized antibodies

    • Solution: Consider antibody sharing within research groups; explore cost-effective options like in-house production for frequently used antibodies

  • Future Directions for Improvement:

    • Development of recombinant antibodies for higher reproducibility

    • More comprehensive validation across multiple applications

    • Generation of knockout-validated antibodies

    • Community-based validation reporting to share experiences

The high cost of antibodies remains a significant challenge in research, particularly in academic settings with limited budgets. As one researcher noted, "We're investing thousands of dollars on small vials of antibodies that may not even function as intended. And if they fail? Tough luck. Just grab another vial, which will set you back another $400."

How does HELLS contribute to germinal center dynamics and long-term humoral immunity?

Recent research has uncovered critical roles for HELLS in germinal center (GC) dynamics and the establishment of long-term humoral immunity:

  • Germinal Center Formation and Maintenance:

    • HELLS-deficient mice can initially form germinal centers but show accelerated decay of GC B cells

    • HELLS expression is essential for the sustained output of germinal centers over time

    • Loss of HELLS leads to premature termination of the GC reaction

  • DNA Methylation Maintenance in Rapidly Dividing GC B Cells:

    • HELLS maintains DNA methylation patterns during rapid proliferation of GC B cells

    • HELLS deficiency induces dramatic DNA hypomethylation

    • This leads to massive de-repression of evolutionary recent retrotransposons

    • Interestingly, this hypomethylation does not directly affect GC B cell survival

  • Cell Fate Determination in the Germinal Center:

    • HELLS-deficient GC B cells prematurely upregulate either:

      • Memory B cell markers, or

      • Transcription factor ATF4 (driving an mTORC1-dependent metabolic program typical of plasma cells)

    • This suggests HELLS regulates proper timing of cell fate decisions in the GC

  • Impact on Antibody Affinity Maturation:

    • HELLS is required for efficient antibody affinity maturation

    • Memory B cells from HELLS-deficient mice show severely reduced high-affinity antibody mutations

    • Specifically, the fraction of memory B cells bearing the W33L or K59R substitutions (which confer increased affinity) is dramatically reduced

  • Mechanism of Action:

    • DNA-methylation maintenance by HELLS appears to be a crucial mechanism to fine-tune the GC transcriptional program

    • Treatment of wild-type mice with a DNMT1-specific inhibitor phenocopies the accelerated kinetics seen in HELLS-deficient mice

    • This confirms the DNA methylation-dependent function of HELLS in GC dynamics

These findings have significant implications for understanding antibody deficiencies in ICF4 syndrome patients and potentially for developing interventions to enhance vaccine efficacy in immunocompromised individuals.

What recent insights have emerged regarding HELLS in transcriptional regulation and disease contexts?

Recent research has revealed expanded roles for HELLS in transcriptional regulation and various disease contexts:

  • Transcriptional Regulation Mechanisms:

    • HELLS directly binds and regulates 467 genes (termed HELLS-direct genes or HDGs)

    • Knockdown of HELLS affects multiple biological processes including T-cell proliferation, JAK/STAT signaling, chromatin organization, and interferon γ signaling

    • HELLS appears to reduce chromatin accessibility at specific loci in T-cell lymphomas

  • Role in DNA Repair Pathways:

    • HELLS facilitates homologous recombination at two-ended DNA breaks

    • It contributes specifically to repair within heterochromatic regions during G2 phase

    • HELLS directly interacts with the end-resection factor CtIP

    • This interaction promotes CtIP accumulation at IR-induced breaks and subsequent end-resection

    • The ATPase activity of HELLS is essential for this function

  • Cancer Implications:

    • HELLS has been implicated in T-cell lymphomas through its regulation of chromatin accessibility

    • Studies show HELLS may control the expansion or survival of lymphoid cells

    • HELLS potentially plays a role in both cancer development and progression

  • Developmental Functions:

    • HELLS is essential for normal development and survival

    • It plays a critical role in the formation and organization of heterochromatin

    • HELLS may control silencing of the imprinted CDKN1C gene through DNA methylation

    • These functions suggest broader roles in developmental gene regulation

  • Emerging Therapeutic Implications:

    • Understanding HELLS function in disease contexts opens new therapeutic possibilities

    • For ICF4 syndrome, addressing the specific B-cell developmental defects could improve outcomes

    • In cancer contexts, targeting HELLS-dependent pathways might offer novel treatment strategies

    • The interaction between HELLS and DNA repair pathways suggests potential roles in modulating therapy responses

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.