HOXD3 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
Typically, we can ship products within 1-3 business days of receiving your order. Delivery times may vary based on the purchasing method or location. Please consult your local distributors for specific delivery time estimates.
Synonyms
Homeo box D3 antibody; Homeobox D3 antibody; Homeobox protein Hox D3 antibody; Homeobox protein Hox-4A antibody; Homeobox protein Hox-D3 antibody; Homeobox protein HoxD3 antibody; Homeodomain protein antibody; HOX 1D antibody; HOX 4 antibody; Hox 4.1 antibody; Hox 4A antibody; HOX D3 antibody; Hox-4.1 mouse homolog of antibody; HOX1D antibody; HOX4 antibody; Hox4.1 antibody; HOX4A antibody; HOXD 3 antibody; HOXD3 antibody; HXD3_HUMAN antibody; MGC10470 antibody
Target Names
HOXD3
Uniprot No.

Target Background

Function
HOXD3 is a sequence-specific transcription factor involved in a developmental regulatory system. It plays a crucial role in establishing the positional identities of cells along the anterior-posterior axis during embryonic development.
Gene References Into Functions
miR-203a
Inhibits hepatocellular carcinoma cell invasion, metastasis, and angiogenesis by negatively targeting HOXD3 and suppressing cell signaling through the VEGFR pathway. PMID: 29402992
EGR1
Acts as a key regulator of miR-203a expression. miR-203a functions as an anti-oncogene to suppress HCC tumorigenesis by targeting HOXD3 through EGFR-related cell signaling pathways. PMID: 27244890
HOXD3
Promotes colorectal cancer cell growth and is essential for the development and survival of malignant human colorectal cancer cells. PMID: 27499213
Promoter hypermethylation of HOXD3 is linked to prostate cancer recurrence. PMID: 24718283
HOXD3 promoter hypermethylation is associated with clinicopathologic features in prostate cancer, particularly in older, higher risk patients. PMID: 24847526
High HOXD3 expression is correlated with invasive breast cancer. PMID: 22935821
Increased promoter methylation levels of HOXD3 are associated with prostate cancer progression. PMID: 21207416
HOXD3 methylation distinguishes low-grade prostate cancers from intermediate and high-grade ones. PMID: 20212450
Transduction of antisense DNA into human melanoma cells results in decreased invasive and motile activities. PMID: 12405287
HOXD3 regulates the expression of integrin alpha5beta1 and integrin alphavbeta3 during angiogenesis in vivo. PMID: 14610084
HOXD3 may potentially improve collagen deposition, angiogenesis, and closure in poorly healing diabetic wounds. PMID: 14633614
HOXD3 may play a significant role in regulating cerebral angiogenesis, and gene transfer of HOXD3 might offer a novel and effective means to stimulate angiogenesis. PMID: 15545924
The karyotype of a patient with Duane syndrome suggests another possible locus for this syndrome. The mapped genes around the deleted region, 1q42.13-43, contain potential candidate genes such as a homeobox gene. PMID: 17126050
Further validation of candidate genes in a separate cohort of low and high-grade prostate cancers using quantitative MethyLight analysis has confirmed DNA hypermethylation of HOXD3 and BMP7. PMID: 19283074
Database Links

HGNC: 5137

OMIM: 142980

KEGG: hsa:3232

STRING: 9606.ENSP00000249440

UniGene: Hs.93574

Protein Families
Antp homeobox family
Subcellular Location
Nucleus.

Q&A

What is HOXD3 and why is it significant in research?

HOXD3 is a sequence-specific transcription factor that belongs to the homeobox gene family, providing cells with specific positional identities during embryonic development. It plays crucial roles in the formation of somitic mesoderm and neural crest-derived structures during development . The significance of HOXD3 extends to pathological conditions, as its dysregulation has been linked to various diseases, including cancer and developmental disorders . Specifically, HOXD3 overexpression in cancer cells has been shown to enhance cell motility, invasion, and metastasis by regulating the expression of adhesion molecules like integrin β3 . This multifaceted role in both development and disease makes HOXD3 a valuable research target for understanding fundamental biological processes and identifying potential therapeutic approaches.

Which applications are most suitable for HOXD3 antibody detection?

HOXD3 antibodies have been validated for multiple applications, with particular strength in Western blot (WB), enzyme-linked immunosorbent assay (ELISA), immunohistochemistry (IHC), and immunofluorescence/immunocytochemistry (IF/ICC) . Western blotting is particularly effective for quantitative analysis of HOXD3 expression levels across different experimental conditions, typically using dilutions of 1:500-1:1000 . For localization studies, IHC (recommended dilution 1:50-1:500) and IF/ICC (recommended dilution 1:200-1:800) provide valuable insights into the nuclear distribution of HOXD3 in tissue sections and cultured cells, respectively . ELISA offers high-throughput screening capabilities at dilutions up to 1:10000 . The choice of application should be guided by specific research questions, with Western blotting being ideal for expression level comparisons, while immunostaining techniques are better suited for localization studies in tissue contexts.

What are the optimal sample preparation methods for HOXD3 detection?

For optimal HOXD3 detection in tissue samples using immunohistochemistry, antigen retrieval with TE buffer at pH 9.0 is recommended, although citrate buffer at pH 6.0 may serve as an alternative . When preparing cell lysates for Western blot analysis, it's critical to include protease inhibitors to prevent degradation of the HOXD3 protein. The nuclear localization of HOXD3 necessitates efficient nuclear extraction protocols for maximum yield . For immunofluorescence studies, cells should be fixed with 4% paraformaldehyde followed by permeabilization with 0.1-0.5% Triton X-100 to ensure nuclear access of the antibody. Regardless of the application, samples should be processed fresh or properly stored at -80°C until use, with repeated freeze-thaw cycles avoided to maintain protein integrity and antibody reactivity.

What are the typical reactivity profiles of commercial HOXD3 antibodies?

Commercial HOXD3 antibodies exhibit varied reactivity profiles across species. Many antibodies, including rabbit polyclonal antibodies, demonstrate reactivity with human and mouse samples . Some antibodies extend their reactivity to include rat samples as well . This cross-reactivity stems from the high conservation of HOXD3 across mammalian species. When selecting an antibody for specific research applications, it's essential to verify that the antibody has been validated in the species of interest. The immunogen sequence used for antibody production significantly influences species reactivity; antibodies raised against highly conserved regions of HOXD3 typically show broader cross-reactivity profiles compared to those targeting more variable regions . Importantly, each antibody should be validated in the researcher's specific experimental system to ensure optimal performance.

How can specificity of HOXD3 antibodies be validated across different experimental models?

Validating HOXD3 antibody specificity requires a multi-pronged approach. First, conduct knockout/knockdown controls where HOXD3 expression is depleted through siRNA, shRNA, or CRISPR-Cas9 techniques, resulting in reduced or absent signal in antibody-based detection methods . Second, perform peptide competition assays by pre-incubating the antibody with the immunizing peptide (such as recombinant HOXD3 protein fragments) before application to samples; a specific antibody will show significantly reduced or eliminated signal . Third, compare results across multiple antibodies targeting different epitopes of HOXD3 to confirm consistent expression patterns. Fourth, employ Western blots to verify that the detected protein has the expected molecular weight of approximately 45-46 kDa . Finally, conduct positive control experiments using cell lines known to express HOXD3, such as U-251MG or HepG2, which have been documented to show reliable HOXD3 expression in validation studies .

What strategies can overcome common challenges in HOXD3 detection by Western blotting?

Western blot detection of HOXD3 presents several challenges that can be addressed through optimized protocols. First, to address non-specific binding, increase blocking stringency using 5% BSA or milk protein and include 0.1% Tween-20 in wash buffers. For weak signal detection, implement signal enhancement techniques such as increasing antibody concentration (1:250-1:500), extending primary antibody incubation to overnight at 4°C, or utilizing high-sensitivity chemiluminescent substrates . When facing multiple bands, enrich nuclear fractions during sample preparation since HOXD3 is primarily localized in the nucleus . For HOXD3 degradation products, add protease inhibitor cocktails to lysis buffers and maintain samples at 4°C throughout processing. If encountering background issues, titrate secondary antibody concentrations down to 1:10,000-1:20,000 and perform more stringent washes between antibody incubations. For molecular weight verification, always include appropriate molecular weight markers to confirm the expected 45-46 kDa band for HOXD3 protein .

What are the critical parameters for successful immunohistochemical detection of HOXD3 in tissue sections?

Successful immunohistochemical detection of HOXD3 in tissue sections depends on several critical parameters. Antigen retrieval represents a crucial step, with TE buffer at pH 9.0 showing superior results for HOXD3 epitope exposure compared to citrate buffer at pH 6.0 . Antibody concentration requires careful titration, typically starting at a 1:50 dilution and increasing to 1:500 depending on tissue type and fixation conditions . Primary antibody incubation should be extended to overnight at 4°C to maximize sensitivity while maintaining specificity. The detection system selection is equally important, with polymer-based systems offering better signal-to-noise ratios than traditional avidin-biotin methods for nuclear transcription factors like HOXD3. Tissue-specific background reduction can be achieved by incorporating species-specific blocking serum (5-10%) during the blocking step. For reproducibility across experiments, standardize all parameters including fixation duration, section thickness (4-6 μm optimal), and counterstaining intensity. Mouse testis tissue has been documented as a reliable positive control for HOXD3 expression validation in IHC applications .

How can researchers optimize double immunofluorescence protocols involving HOXD3 antibodies?

Optimizing double immunofluorescence protocols with HOXD3 antibodies requires careful consideration of several technical aspects. Primary antibody compatibility should be ensured by selecting antibodies raised in different host species (e.g., rabbit anti-HOXD3 paired with mouse antibodies against other targets) to enable selective secondary antibody detection . Sequential staining may be necessary, applying and detecting the HOXD3 antibody first (dilution 1:200-1:800), followed by complete washing and blocking steps before applying the second primary antibody . Signal enhancement through tyramide signal amplification can improve detection of low-abundance HOXD3 protein while maintaining specificity. When analyzing co-localization, spectral unmixing should be employed to eliminate potential bleed-through between fluorescent channels. For nuclear HOXD3 detection alongside cytoplasmic or membrane markers, confocal microscopy with Z-stack acquisition is recommended to accurately distinguish subcellular localization patterns. U2OS cells serve as excellent positive controls for immunofluorescence optimization, as they consistently show nuclear HOXD3 expression .

How should researchers interpret variations in HOXD3 expression levels across different cell types?

Interpreting variations in HOXD3 expression across different cell types requires contextual understanding of its biological functions. HOXD3 expression is developmentally regulated and tissue-specific, with higher expression typically observed in cells undergoing morphogenesis or differentiation . In cancer cells, elevated HOXD3 expression often correlates with increased motility and metastatic potential, as demonstrated in lung cancer A549 cells . When quantifying expression differences, researchers should normalize HOXD3 levels to appropriate housekeeping genes and include positive control samples such as U-251MG or HepG2 cells that reliably express HOXD3 . Temporal expression patterns should also be considered, as HOXD3 may be transiently expressed during specific developmental windows or cellular responses. For comparative analyses across cell types, standardized protein extraction protocols are essential to ensure that differences reflect true biological variation rather than methodological discrepancies. Finally, functional validation through gain-of-function or loss-of-function experiments should complement expression data to establish biological significance of observed variations.

What is the relationship between HOXD3 expression and integrin signaling in experimental models?

The relationship between HOXD3 expression and integrin signaling represents a critical axis in cell adhesion and motility regulation. HOXD3 overexpression directly upregulates integrin β3 expression by binding to its promoter region, subsequently forming active αvβ3 integrin heterodimers that enhance cell-extracellular matrix interactions . This HOXD3-induced integrin β3 upregulation significantly increases cell motility and invasion capabilities, as demonstrated in lung cancer A549 cells and human endothelial cells . Additionally, HOXD3 overexpression may simultaneously downregulate E-cadherin expression, further promoting cell dissociation and migration . When analyzing this relationship experimentally, researchers should assess both HOXD3 and integrin β3 expression levels concurrently, potentially using dual immunofluorescence techniques. The functional consequences can be evaluated through various motility assays, including haptotaxis, phagokinetic track assays, and wound-healing assays, which have demonstrated that HOXD3-transfected cells exhibit higher migratory activity than control cells . To establish a causal relationship, targeted inhibition of integrin β3 in HOXD3-overexpressing cells should reverse the enhanced motility phenotype if the relationship is mechanistically linked.

How can researchers differentiate between specific and non-specific binding in HOXD3 antibody applications?

Differentiating between specific and non-specific binding in HOXD3 antibody applications requires systematic validation strategies. Peptide competition assays provide direct evidence of binding specificity, where pre-incubation of the antibody with the immunizing peptide should substantially reduce or eliminate specific signals while leaving non-specific binding unaffected . The molecular weight verification in Western blot applications should confirm detection at the expected 45-46 kDa position corresponding to HOXD3 protein . Genetic knockdown experiments using siRNA or CRISPR-Cas9 techniques targeting HOXD3 should result in corresponding signal reduction in proportion to the knockdown efficiency. The subcellular localization pattern should align with HOXD3's known nuclear distribution; cytoplasmic signals may indicate non-specific binding unless validated through additional methods . Multiple antibody validation involves comparing staining patterns from different antibodies targeting distinct HOXD3 epitopes; concordant results suggest specific detection. Proper controls in each experiment must include no-primary-antibody controls to assess secondary antibody background, isotype controls to evaluate non-specific binding, and positive/negative tissue controls (such as mouse testis as a positive control for IHC applications) .

What factors should be considered when comparing HOXD3 expression data across different detection methods?

When comparing HOXD3 expression data across different detection methods, researchers must consider several methodological factors that influence results interpretation. Sensitivity differences exist between techniques, with Western blotting providing quantitative expression data, ELISA offering high-throughput screening capabilities, and immunostaining methods revealing spatial distribution . Sample preparation variations significantly impact results; for instance, nuclear extraction efficiency directly affects HOXD3 detection levels in Western blots due to its nuclear localization . Antibody performance may vary across applications, with some antibodies performing optimally in Western blots but poorly in IHC or vice versa, necessitating application-specific validation . Signal amplification methods differ between techniques, with immunofluorescence potentially requiring signal enhancement for optimal HOXD3 visualization. Quantification approaches vary fundamentally, from densitometry in Western blots to intensity measurements in immunofluorescence, making direct numerical comparisons challenging. When interpreting discrepancies between methods, researchers should consider these technical variations and ideally corroborate findings using complementary approaches, such as validating protein expression changes with corresponding mRNA level measurements through qRT-PCR.

How can HOXD3 antibodies be utilized to study its role in cancer progression and metastasis?

HOXD3 antibodies serve as valuable tools for investigating its role in cancer progression through multiple experimental approaches. Immunohistochemical profiling of HOXD3 expression across tumor stages and grades can establish correlations between expression levels and prognostic outcomes, using antibody dilutions of 1:50-1:500 for tissue microarray analysis . Co-immunofluorescence studies combining HOXD3 (1:200-1:800 dilution) with metastasis markers like integrin αvβ3 can reveal spatial relationships between HOXD3 expression and invasive phenotypes . Western blot analysis (1:500-1:1000 dilution) of HOXD3 expression following treatments with potential anti-metastatic compounds helps identify regulatory mechanisms and therapeutic opportunities . Chromatin immunoprecipitation (ChIP) using HOXD3 antibodies can map its direct transcriptional targets, particularly focusing on adhesion molecules like integrin β3 whose promoter contains HOXD3 binding sites . For functional validation, HOXD3 expression can be modulated through transfection approaches, followed by motility assays like wound-healing or transwell migration assays to establish causative relationships between HOXD3 levels and invasive behavior, as demonstrated in A549 lung cancer cells where HOXD3-overexpression significantly enhanced motility compared to control cells .

What are the best practices for using HOXD3 antibodies in developmental biology research?

In developmental biology research, HOXD3 antibody applications require specific methodological considerations. Embryonic tissue preparation demands careful fixation optimization, typically using 4% paraformaldehyde for 24-48 hours depending on embryonic stage, followed by proper processing to preserve antigenicity while maintaining morphology. For temporal expression analysis, consistent sampling across developmental timepoints is crucial, with immunohistochemistry dilutions of 1:50-1:200 typically providing optimal signal-to-noise ratios in embryonic tissues . Multi-color immunofluorescence (1:200-1:800 dilution) enables co-localization studies with developmental markers to establish relationships between HOXD3 expression and specific differentiation events . When examining HOXD3's role in neural crest and somitic mesoderm development, whole-mount immunohistochemistry may be employed, though this requires extensive optimization of antibody penetration through embryonic tissues . For quantitative developmental comparisons, consistent imaging parameters must be maintained across specimens, with z-stack confocal microscopy recommended for three-dimensional tissue analysis. Validation of antibody specificity is particularly important in developmental studies where cross-reactivity with other HOX family members may occur due to sequence homology; knockout embryos serve as ideal negative controls when available.

How can researchers effectively use HOXD3 antibodies to investigate gene regulation mechanisms?

Investigating gene regulation mechanisms involving HOXD3 requires specialized antibody applications focusing on transcriptional activity. Chromatin immunoprecipitation (ChIP) assays using HOXD3 antibodies can identify direct binding sites on target gene promoters, such as the documented interaction between HOXD3 and the integrin β3 promoter region . For this application, antibodies must be validated for specific immunoprecipitation efficiency and typically require 2-5 μg per ChIP reaction. Sequential ChIP (ChIP-reChIP) can reveal co-occupancy of HOXD3 with cofactors or other transcription factors on regulatory elements. Combining ChIP with high-throughput sequencing (ChIP-seq) enables genome-wide mapping of HOXD3 binding sites, revealing previously unknown regulatory targets. Co-immunoprecipitation studies using HOXD3 antibodies can identify protein-protein interactions that modulate its transcriptional activity. For analyzing HOXD3 nuclear localization dynamics in response to signaling events, time-course immunofluorescence studies (1:200-1:800 dilution) following specific stimuli can be employed . When examining post-translational modifications that affect HOXD3 activity, specialized antibodies recognizing specific modifications may complement standard HOXD3 detection. These approaches collectively provide mechanistic insights into how HOXD3 regulates target genes like integrin β3, contributing to phenotypes such as enhanced cell motility in cancer models .

What experimental design considerations are important when using HOXD3 antibodies in tissue-specific studies?

Tissue-specific studies utilizing HOXD3 antibodies require careful experimental design considerations to ensure valid and reproducible results. Comparative expression analysis across tissues should include standardized controls, as HOXD3 expression varies naturally between tissue types with documented expression in testis tissue serving as a reliable positive control for immunohistochemical applications . Antigen retrieval method selection significantly impacts detection sensitivity in different tissues, with TE buffer at pH 9.0 showing superior results for HOXD3 epitope retrieval in most tissue types, though optimization may be required for specific tissues . When examining co-expression with tissue-specific markers, sequential staining protocols may be necessary to avoid antibody cross-reactivity issues. For quantitative comparisons across tissues with varying HOXD3 expression levels, calibration curves using recombinant HOXD3 protein can establish absolute quantification parameters. The antibody penetration efficiency varies between tissue types due to differences in density and composition, potentially requiring adjusted incubation times or concentration for optimal results. Background autofluorescence, particularly in tissues like liver or kidney, necessitates appropriate quenching procedures for immunofluorescence applications. Finally, when interpreting tissue-specific HOXD3 expression patterns, developmental stage and physiological state must be considered as contextual factors that influence expression levels and localization patterns.

What are the key differences between monoclonal and polyclonal HOXD3 antibodies in research applications?

Polyclonal HOXD3 antibodies, such as the rabbit polyclonal antibodies described in the search results, recognize multiple epitopes on the HOXD3 protein, offering higher sensitivity but potentially increased background compared to monoclonal alternatives . This multi-epitope recognition provides advantages in applications where protein conformation may be altered, such as formalin-fixed paraffin-embedded tissues in immunohistochemistry, where polyclonal antibodies consistently demonstrate robust detection capability . The production methods differ significantly: polyclonal antibodies are generated by immunizing animals (typically rabbits) with synthetic peptides or recombinant protein fragments corresponding to specific regions of HOXD3, such as amino acids 211-260 or 263-432 of human HOXD3 . These antibodies typically undergo affinity purification to enhance specificity, as documented in the preparation of commercial antibodies . For applications requiring quantitative comparison across experiments, monoclonal antibodies would theoretically provide more consistent lot-to-lot reproducibility, though well-characterized polyclonal antibodies with defined immunogens, such as those targeting specific amino acid sequences of HOXD3, can also deliver reliable results when properly validated in each experimental system .

What storage and handling practices ensure optimal performance of HOXD3 antibodies?

Optimal performance of HOXD3 antibodies depends on proper storage and handling practices throughout their lifecycle. Upon receipt, antibodies should be aliquoted into single-use volumes to minimize freeze-thaw cycles, which can significantly degrade antibody performance over time . Storage temperature requirements indicate that while shipping can occur at 4°C, long-term storage should be at -20°C, with glycerol-containing formulations (typically 50% glycerol) preventing freeze-thaw damage . The buffer composition plays a crucial role in stability, with most commercial HOXD3 antibodies supplied in Phosphate Buffered Saline (without Mg²⁺ and Ca²⁺) at pH 7.4, containing 150mM NaCl, 0.02% Sodium Azide, and 50% Glycerol . Before use, antibodies should be completely thawed and gently mixed to ensure homogeneity without introducing damaging bubbles. Working dilutions should be prepared fresh for each experiment using high-quality diluents compatible with the intended application, typically containing 1-5% BSA or normal serum from the same species as the secondary antibody to reduce background. Once diluted, antibodies should be used immediately or stored at 4°C for no more than 1-2 weeks to maintain optimal reactivity and specificity .

How do researchers evaluate batch-to-batch consistency of HOXD3 antibodies?

Evaluating batch-to-batch consistency of HOXD3 antibodies requires systematic quality control procedures to ensure reproducible experimental results. Internal reference standards should be established by preserving a portion of a well-characterized antibody lot to serve as a benchmark for comparison with new batches. Parallel testing protocols involve running side-by-side comparisons of new and reference batches across multiple applications, including Western blot (1:500-1:1000 dilution), immunohistochemistry (1:50-1:500 dilution), and immunofluorescence (1:200-1:800 dilution) as applicable . Quantitative metrics for comparison include signal intensity at standardized exposure settings, signal-to-noise ratio, and detection sensitivity with serially diluted sample loads. Epitope mapping confirmation ensures that new batches recognize the same regions of the HOXD3 protein as the reference batch, particularly important for polyclonal antibodies where epitope drift may occur between productions. Consistent positive control samples must be included in validation studies, with documented HOXD3-expressing cell lines such as U-251MG, HepG2, or U2OS serving as reliable standards . Finally, documentation practices should include detailed records of validation results with standardized imaging or detection parameters to facilitate objective comparisons between batches and ensure experimental reproducibility across studies.

What validation data should researchers expect from manufacturers of HOXD3 antibodies?

Comprehensive validation data from HOXD3 antibody manufacturers should include several key components to ensure research reliability. Application-specific validation results should demonstrate performance in each claimed application (Western blot, IHC, IF/ICC, ELISA) with representative images and recommended dilution ranges for each technique (e.g., 1:500-1:1000 for WB, 1:50-1:500 for IHC, 1:200-1:800 for IF/ICC) . Specificity documentation should include Western blot images showing detection of the expected 45-46 kDa HOXD3 protein band in positive control lysates, such as from HepG2 cells . Reactivity profiling across species should clearly indicate which species have been experimentally validated (typically human and mouse for HOXD3 antibodies) versus predicted reactivity based on sequence homology . Immunogen information should detail the specific sequence used for antibody generation, such as "amino acids 211-260 of human HOXD3" or "recombinant fusion protein containing amino acids 263-432 of human HOXD3" . Positive control recommendations should specify cell lines or tissues known to express HOXD3, such as U-251MG, HepG2, or mouse testis tissue . Finally, protocol recommendations should include detailed application-specific guidance, including antigen retrieval methods for IHC (such as TE buffer pH 9.0 or citrate buffer pH 6.0), buffer compositions, and optimal incubation parameters .

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.