The HECTD1 antibody is a specialized immunological tool designed to detect and study the HECT domain E3 ubiquitin-protein ligase 1 (HECTD1), a critical regulator of cellular processes ranging from ribosome assembly to cancer progression . This antibody enables researchers to investigate HECTD1's expression patterns, molecular interactions, and functional roles through techniques like Western blotting, immunofluorescence, and immunohistochemistry .
HECTD1 antibodies have been instrumental in:
Identifying subcellular localization: Demonstrated nuclear-cytoplasmic shuttling in human cells via immunofluorescence .
Quantifying protein expression: Used to measure HECTD1 levels in breast cancer tissues, revealing prognostic significance .
Mechanistic studies: Validated HECTD1's role in ubiquitinating ribosomal assembly factors (e.g., ZNF622) and regulating mitosis .
| Parameter | Detail |
|---|---|
| Host Species | Rabbit (polyclonal) |
| Reactivity | Human, mouse |
| Applications | WB (1:200–1:1000), IF (1:500), IHC (1:100) |
| Validation | Knockdown/WT comparisons in HEK293T, TF-1, and primary HSCs |
Critical validation steps include:
Specificity confirmation via CRISPR-Cas9 knockout lines showing complete signal loss
Quantitative correlation between mRNA levels (RNA-seq) and protein detection (r = 0.79, P<0.001)
Oncogenic role: HECTD1 maintains ER-α transcriptional activity by degrading RIP40 corepressor (K48-linked ubiquitination) .
Therapeutic target potential: Breast cancers with HECTD1 H-scores <100 show 2.1x higher metastasis risk (P=0.009) .
Developmental impact: Hectd1<sup>-/-</sup> murine HSCs exhibit 60% reduced repopulation capacity due to ribosomal 60S/40S joining defects .
HECTD1 antibodies have been successfully tested in multiple applications including:
For optimal results, antibody dilutions should be empirically determined for each experimental system as sensitivity may vary based on protein expression levels and sample preparation methods.
For immunofluorescence detection of HECTD1:
Seed cells into 24-well plates containing 12-mm round cover glass slips
Prior to treatment, serum-starve cells for 16 hours
Fix cells with 4% formaldehyde for 15 minutes at room temperature
Permeabilize with 0.15% Triton-X100/PBS for 15 minutes
Block with 5% BSA/PBS for 1 hour at room temperature
Incubate with anti-HECTD1 primary antibody (1:200 dilution) overnight at 4°C
Wash with PBS (3 times, 15 minutes each)
Incubate with Alexa Fluor-conjugated secondary antibodies (1:500) for 1 hour
Stain nuclei with DAPI for 3 minutes
Mount with anti-fade mountant and visualize using confocal microscopy
For IHC applications, antigen retrieval with TE buffer pH 9.0 is recommended, though citrate buffer pH 6.0 can be used as an alternative .
HECTD1 is broadly expressed across multiple tissue types:
HECTD1 expresses broadly in nervous system, cardiovascular tissues, and epithelial cells, making these tissues suitable for positive controls in antibody validation studies .
A multi-step validation approach is recommended:
Cell line selection: Utilize established cell models such as HEK293T, HeLa, or TF-1 cells that reliably express HECTD1
Knockout/knockdown strategies:
Validation experiments:
Western blot analysis comparing wild-type and knockout/knockdown samples
Include antibody specificity controls such as the HECTD1 XC mouse line where a gene trap insertion disrupts the C-terminal domain, allowing for negative control validation
For transfected samples, include both wild-type HECTD1 and ligase-deficient HECTD1 C2579G constructs
Researchers should note that knockout validation provides more definitive evidence of antibody specificity than knockdown approaches, as residual protein can complicate interpretation in knockdown systems .
For co-immunoprecipitation of endogenous HECTD1 and its interaction partners:
Protocol for stringent conditions (endogenous proteins):
Prepare tissue/cell lysates in RIPA buffer
Incubate lysate with anti-HECTD1 antibody overnight at 4°C
Add protein A/G beads and incubate for 2-4 hours
Wash beads 4-5 times with RIPA buffer
Elute bound proteins with SDS sample buffer and analyze by western blotting
Protocol for overexpressed proteins:
Transfect cells with tagged HECTD1 constructs (e.g., HA-HECTD1) and potential interacting partners (e.g., Myc-RARA)
Lyse cells in appropriate buffer (less stringent than RIPA)
Immunoprecipitate using anti-tag antibodies (e.g., anti-HA agarose beads)
Perform specific elution with HA peptide to minimize non-specific binding
Verify elution efficiency with glycine elution as a secondary step
When studying HECTD1 interactions, it's critical to include appropriate controls:
IgG control antibody
Ligase-deficient HECTD1 mutant (C2579G) to distinguish between substrate binding and catalytic activity
Empty vector controls for tagged protein experiments
To analyze HECTD1-mediated ubiquitination of specific substrates:
Experimental design approach:
Co-transfect cells with Myc-tagged substrate (e.g., Myc-RARA), HA-HECTD1 (wild-type), and ligase-deficient HECTD1 C2579G
Treat cells with proteasome inhibitor (e.g., ALLN) 4 hours before lysis to prevent degradation of ubiquitinated proteins
Immunoprecipitate substrate with anti-Myc antibody
Detect ubiquitination by western blotting with anti-ubiquitin antibodies
Ubiquitin chain-specific detection:
Controls for ubiquitination assays:
HECTD1 has been reported to assemble both proteasomal (K48-linked) and non-proteasomal (K63-linked) ubiquitin signals, so experimental approaches should be capable of distinguishing these outcomes .
HECTD1 detection presents several challenges due to its large size (~290 kDa) and potential for degradation:
Size-related issues:
Protein degradation concerns:
Signal optimization:
Recommended antibody dilutions for western blotting range from 1:500 to 1:3000
Normalize loading with appropriate controls (GAPDH is commonly used)
Extended blocking (2 hours or overnight) may reduce background
If detecting endogenous HECTD1, consider longer exposure times due to potentially low expression levels
Verification strategies:
HECTD1 contains multiple functional domains that contribute to its diverse cellular roles:
Domain-specific analysis approaches:
Functional domain analysis using mutant models:
The HECTD1 XC mouse line expresses a truncated HECTD1 protein retaining substrate binding domains but with a disrupted C-terminal ubiquitin ligase domain
The HECTD1 opm mouse line harbors an ENU-induced nonsense mutation that truncates the 2610-amino-acid HECTD1 protein after amino acid 144
These models allow separation of binding function from catalytic activity
Detecting domain-specific interactions:
For subcellular localization analysis, use prediction software such as RSLpred and PSORT II
When studying nuclear-cytoplasmic shuttling, treatments with nuclear export inhibitors like Leptomycin B (LMB, 50 nM) or ivermectin (IVE, 1 μM) can help determine functional domain requirements
Perform domain mapping through co-immunoprecipitation with truncated constructs
Researchers should note that different functional domains may contribute to distinct cellular processes, with the HECT domain being critical for ubiquitin ligase activity but not necessarily for protein-protein interactions .
Based on established research findings, HECTD1 contributes to cell proliferation through regulation of mitosis:
Cellular model selection:
Experimental design framework:
Cell proliferation assays:
Mitotic phase analysis:
Spindle assembly checkpoint evaluation:
Critical controls:
Include both wild-type and ligase-deficient HECTD1 constructs in rescue experiments
Compare multiple HECTD1 knockout/knockdown clones to rule out off-target effects
Verify mitotic timing alterations with multiple methodological approaches
HECTD1 depletion has been shown to increase the proportion of cells with aligned chromosomes and extend the time from NEBD to anaphase onset, suggesting a role in mitotic progression regulation .
HECTD1 plays a critical role in hematopoietic stem cell (HSC) function and regeneration:
In vivo experimental approaches:
Competitive bone marrow transplantation assay:
Inject bone marrow cells from HECTD1-deficient donor mice (CD45.2) with equal numbers of competitor cells (CD45.1) into lethally irradiated recipients
Analyze donor chimerism in peripheral blood by flow cytometry every 4 weeks
Determine HSC frequencies using extreme limiting dilution analysis (ELDA)
Purified HSC transplantation:
Isolate HSCs (LSK CD150+CD48−) by flow cytometric sorting
Transplant defined numbers of cells into lethally irradiated recipients with competitors
Compare reconstitution ability between wild-type and HECTD1-deficient HSCs
This approach allows distinction between intrinsic HSC defects and microenvironment effects
Ex vivo culture systems:
Human cell line models:
Research has demonstrated that HECTD1 deficiency decreases both HSC frequency and function in vivo, with particularly pronounced effects on HSC maintenance during ex vivo culture .
HECTD1 regulates multiple substrates through ubiquitination, including RARA (retinoic acid receptor alpha) and SNAIL:
Substrate-specific detection methods:
RARA interaction studies:
Co-immunoprecipitate HECTD1 and RARA under various conditions
Analyze ubiquitination patterns using FK1 (polyubiquitin-specific) and FK2 (mono- and polyubiquitin) antibodies
Compare wild-type and ligase-deficient HECTD1 effects on RARA stability and ubiquitination
Assess the impact on retinoic acid signaling using reporter assays
SNAIL regulation analysis:
Functional outcome assessment:
For RARA-mediated pathways:
For SNAIL-mediated processes:
Critical controls for substrate studies:
Research has shown that HECTD1 interacts with RARA and influences its ubiquitination state, with HECTD1 deficiency leading to reduced retinoic acid signaling in embryos. Similarly, HECTD1 regulates SNAIL stability through ubiquitination, with HECTD1 knockdown increasing SNAIL expression levels .
Recent research has identified HECTD1 as both a regulator and a substrate in apoptotic pathways:
Experimental design for HECTD1 cleavage studies:
Induce apoptosis through intrinsic or extrinsic pathways
Monitor HECTD1 cleavage using antibodies that can detect both full-length and cleaved fragments
Verify cleavage site identification through mutagenesis
Use domain-swapping mutagenesis (e.g., fusing ubiquitin to the N-terminus of C-terminal HECTD1 fragments) to study fragment stability
Caspase-mediated regulation:
Technical considerations:
Ensure sufficient separation of high molecular weight proteins when analyzing HECTD1 cleavage
Include appropriate controls for apoptosis induction
Consider temporal dynamics of HECTD1 cleavage in relation to other apoptotic events
Analyze subcellular localization changes of HECTD1 fragments after cleavage
Research has demonstrated that HECTD1 is cleaved during apoptosis and may serve as both a regulator and substrate of caspase-3, suggesting a complex role in cell death pathways .
Mass spectrometry (MS) provides powerful tools for HECTD1 research:
Identification of interaction partners:
Perform affinity purification of HECTD1 complexes followed by MS
Express HA-tagged HECTD1 in appropriate cell systems
Use HA peptide affinity elution to minimize non-specific binding
Analyze results using the Contaminant Repository for Affinity Purification (CRAPome) to filter out common contaminants
Validate MS-identified interactions through orthogonal methods
Ubiquitination site mapping:
Identify specific lysine residues on substrates that are ubiquitinated by HECTD1
Distinguish between different ubiquitin chain types (K48, K63, K29)
Quantify changes in ubiquitination patterns upon HECTD1 manipulation
Technical considerations:
Perform experiments in triplicate to ensure reproducibility
Include appropriate negative controls (vector alone) for comparison
Verify efficiency of purification through sequential elution steps
Consider using SILAC or TMT labeling for quantitative analysis of HECTD1-dependent changes
This approach has successfully identified HECTD1 interaction partners in hematopoietic cells, revealing connections to ribosome assembly factors like ZNF622 .