HOXA11 Antibody, Biotin conjugated

Shipped with Ice Packs
In Stock

Description

HOXA11 Antibody: General Characteristics and Conjugation Options

HOXA11 antibodies are used to detect and study the protein’s role in developmental biology, oncology, and reproductive health. Key features include:

Conjugate TypeExamplesApplicationsSources
HRPsc-393440 HRP (Santa Cruz)Western blot, ELISA
Fluorophores (FITC/PE)sc-393440 FITC/PE (Santa Cruz)Immunofluorescence, flow cytometry
Alexa Fluor®sc-393440 AF488/AF647 (Santa Cruz)High-resolution imaging, multiplex assays
Agarosesc-393440 AC (Santa Cruz)Immunoprecipitation, affinity purification

Biotin Conjugation: Technical Considerations

Biotinylated antibodies leverage the high-affinity biotin-streptavidin interaction for sensitive detection in assays like ELISA, IHC, and ChIP. Key steps include:

  1. Antibody Preparation: Removal of stabilizers (e.g., BSA, sodium azide) to avoid interference .

  2. Conjugation Chemistry: Use of NHS-biotin or other biotinylation kits under controlled pH and temperature .

  3. Buffer Optimization: Storage in PBS with cryoprotectants (e.g., glycerol) to maintain stability .

Example Protocol (Adapted from Bosterbio Q&A ):

  1. Purify Antibody: Use desalting columns to remove sodium azide/BSA.

  2. Biotinylation: React with NHS-biotin (1:1 molar ratio).

  3. Dialyze: Remove excess biotin.

  4. Store: In PBS + 50% glycerol at -20°C.

Research Applications of HOXA11 Antibodies

HOXA11 antibodies are pivotal in studying its role in:

Developmental Biology

  • Embryogenesis: Regulates uterus formation and endometrial cycling .

  • Neurogenesis: Involved in CNS and limb development .

Oncology

  • Glioma: HOXA11-AS (antisense RNA) promotes malignancy via β-catenin/c-Myc and ROS resistance pathways .

  • AML: HOXA11 expression correlates with Ara-C sensitivity and apoptosis .

  • HCC: HOXA11-AS knockdown inhibits Wnt signaling and stem cell maintenance .

Reproductive Health

  • Endometrial Function: Essential for blastocyst implantation during the menstrual cycle .

Challenges and Optimization Strategies

  1. Biotin Interference: High-biotin samples may saturate streptavidin, requiring buffer optimization .

  2. Conjugation Efficiency: Excess biotin can cause aggregation; titration is critical .

  3. Cross-Reactivity: Ensure antibodies do not bind non-target epitopes (e.g., HOXD11 in polyclonal antisera) .

Future Directions

  • Therapeutic Targets: HOXA11-AS silencing in glioma and HCC highlights its potential as a drug target .

  • Multiplex Assays: Biotin-conjugated HOXA11 antibodies could enable simultaneous detection with other biomarkers.

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship the 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 estimates.
Synonyms
Homeo box 1I antibody; Homeo box A11 antibody; Homeobox A11 antibody; Homeobox protein Hox A11 antibody; Homeobox protein Hox-1I antibody; Homeobox protein Hox-A11 antibody; Homeobox protein HOXA11 antibody; Hox 1I antibody; HOX1 antibody; HOX1I antibody; HOXA11 antibody; HXA11_HUMAN antibody
Target Names
HOXA11
Uniprot No.

Target Background

Function
HOXA11 is a sequence-specific transcription factor that plays a vital role in a developmental regulatory system. This system provides cells with specific positional identities along the anterior-posterior axis, influencing their developmental fate.
Gene References Into Functions
  1. Research has uncovered a potential regulatory mechanism between the long non-coding RNA HOTTIP and the HOXA11 gene. This suggests that HOTTIP might influence the expression of HOXA11, which is known to be involved in cell growth, migration, and apoptosis in breast cancer MCF-7 cells. PMID: 29415429
  2. Elevated expression of the lncRNA HOXA11-AS has been linked to increased breast cancer invasion and metastasis. This is attributed to its influence on the epithelial-mesenchymal transition (EMT). PMID: 28701685
  3. Silencing of HOXA11 has been shown to suppress the proliferation and invasion of osteosarcoma cells, while inducing cell cycle arrest. PMID: 28558357
  4. Studies have shown that aberrant hypermethylation and subsequent downregulation of HOXA11 may contribute to the progression of lung adenocarcinoma. PMID: 28380439
  5. Hypermethylation of HOXA11 has been identified as an independent prognostic biomarker in breast cancer. This suggests that HOXA11 might act as a potential tumor suppressor. PMID: 28038461
  6. Underexpression of HOXA11 has been linked to treatment resistance and poor prognosis in glioblastoma multiforme (GBM). PMID: 27456940
  7. While mutations in the coding sequence of HOXA11 are not commonly associated with cryptorchidism, common polymorphisms in the HOXA11 gene might contribute to the risk of developing unilateral cryptorchidism. PMID: 28261830
  8. Research has demonstrated that HOXA11-antisense (AS) acts as a molecular sponge for miR-124. Overexpression of miR-124 attenuates the proliferation and invasion-promoting effects of HOXA11-AS. PMID: 28749709
  9. Bioinformatics predictions and luciferase reporter assays have confirmed that miR-140-5p directly targets HOXA11-antisense RNA at its 3'-UTR. PMID: 28832185
  10. HOXA11-AS functions as a competing endogenous RNA, influencing gene expression by competing for microRNA binding sites. PMID: 29034803
  11. Studies have shown that HOXA11 acts as a tumor suppressor in renal cell carcinoma. PMID: 28423531
  12. Laparoscopic endometrioma resection has been found to increase peri-implantation endometrial HOXA-11 mRNA expression. PMID: 26056923
  13. Epigenetic silencing of HOXA11 promotes gastric cancer proliferation, migration, and invasion through the activation of Wnt signaling. PMID: 25590359
  14. The overexpression of miR-30d or 181a has been shown to suppress HOXA11 mRNA expression. PMID: 25630974
  15. Low HOXA11 expression has been associated with hepatocellular carcinoma. PMID: 24306662
  16. Mutations in the coding region of HOXA11 are not commonly observed in Chinese women with Mullerian duct anomalies. PMID: 24745830
  17. Research suggests that HOXA11 hypermethylation may contribute to the progression of non-small cell lung cancer (NSCLC) by promoting cell proliferation or migration. PMID: 24259349
  18. Lower expression of HOXA11 has been linked to endometriosis-associated infertility. PMID: 23979130
  19. Promoter methylation of HOXA11 is associated with glioblastoma. PMID: 23624749
  20. Reduced HOXA11 expression may contribute to endometriosis-associated infertility. PMID: 22233680
  21. Endometrial HOXA11 and LIF mRNA expression levels (normalized to beta-actin expression) were significantly decreased in the endometrium of infertile patients with endometriosis compared with healthy fertile controls. PMID: 21987111
  22. Studies indicate that HOXA-11 gene expression in the endometrium during the implantation window may not be altered in patients with idiopathic infertility. PMID: 21526497
  23. Expression of HOXA11 has been shown to repress the expression of p53 in uterosacral ligaments. PMID: 19372592
  24. Findings suggest that altered HOXA11 gene expression in the endometrium during a menstrual cycle may be a common occurrence among patients with endometriosis and may contribute to infertility in this population. PMID: 20695189
  25. A study revealed that NUP98 was fused in-frame to HOXA11 in a patient with chronic myelogenous leukemia (CML). PMID: 11830496
  26. HOXA11 is expressed in the developing kidney. PMID: 12060755
  27. HOXA gene expression is upregulated at the ectopic implantation site in the fallopian tube, suggesting its potential role in ectopic pregnancies. PMID: 15167850
  28. Research has revealed a novel expression pattern for the homeobox A11 (HOXA11) gene in the human endometrium. PMID: 15467538
  29. The HOXA11 gene cluster is involved in T-cell malignancies, leading to deregulated HOXA gene expression. This is consistent with previous reports suggesting HOXA deregulation in MLL-rearranged T cell lymphoblastic leukemia. PMID: 15674412
  30. HOXA11 is critical for the development and maintenance of uterosacral ligaments and its deficiency is associated with pelvic prolapse. PMID: 18274672
  31. Findings do not support the hypothesis that mutations in the HOXA11 coding regions are involved in the pathogenesis of human non-syndromal congenital renal parenchymal malformations. PMID: 19255789
  32. Methylation of the HOXA11 gene is associated with high-grade squamous intraepithelial lesions in cervical cancer. PMID: 19609949

Show More

Hide All

Database Links

HGNC: 5101

OMIM: 142958

KEGG: hsa:3207

STRING: 9606.ENSP00000006015

UniGene: Hs.249171

Involvement In Disease
Radioulnar synostosis with amegakaryocytic thrombocytopenia 1 (RUSAT1)
Protein Families
Abd-B homeobox family
Subcellular Location
Nucleus.

Q&A

What is HOXA11 and why is it a significant research target?

HOXA11 is a member of the AbdB homeobox family that functions as a sequence-specific transcription factor. It plays a crucial role in regulating embryonic development by providing cells with specific positional identities on the anterior-posterior axis . HOXA11 is essential for female fertility as it governs the cyclic development of the adult endometrium and the formation of the uterus during embryogenesis . Its expression significantly increases during the mid-luteal phase of the menstrual cycle to facilitate blastocyst implantation, highlighting its importance in reproductive biology . Additionally, HOXA11 interacts with various transcription factors and signaling pathways vital for proper morphogenesis, making it a key player in the development of the vertebrate central nervous system, heart, axial skeleton, limbs, gut, and urogenital tract . Due to these critical developmental roles, HOXA11 antibodies are valuable tools for studying embryonic development, reproductive biology, and morphogenesis.

What are the primary applications of biotin-conjugated HOXA11 antibodies in research?

Biotin-conjugated HOXA11 antibodies are versatile tools applicable across multiple experimental techniques. Based on antibody characteristics similar to those seen with biotin-conjugated HOX family antibodies, these reagents can be effectively utilized in western blotting (WB), immunoprecipitation (IP), immunofluorescence (IF), immunohistochemistry (IHC), flow cytometry (FC), and enzyme-linked immunosorbent assay (ELISA) . The biotin conjugation provides significant advantages for detection systems utilizing avidin or streptavidin complexes, allowing for signal amplification and increased sensitivity. For flow cytometry applications, biotin-conjugated antibodies typically use dilutions around 1:100, while for immunofluorescence, similar dilutions are recommended . Western blotting applications generally require more dilute preparations (1:500-2000), and immunohistochemistry protocols typically use intermediate dilutions (approximately 1:150) . The versatility of biotin-conjugated antibodies makes them particularly valuable for multicolor immunofluorescence studies and chromatin immunoprecipitation experiments where signal enhancement is crucial.

How does the structure of HOXA11 protein affect antibody recognition?

The HOXA11 protein contains specific structural domains that influence antibody binding and recognition. Based on western blot data from commercial antibodies, HOXA11 has a predicted molecular weight of approximately 34 kDa, though it may appear at around 30 kDa on gels due to post-translational modifications or protein folding . When developing immunodetection protocols, researchers should consider that HOXA11 contains a homeodomain that is highly conserved among HOX family proteins, which may affect antibody specificity. The use of synthetic peptide immunogens corresponding to unique regions of HOXA11 helps ensure specificity when distinguishing between closely related HOX proteins . Epitope accessibility can be affected by protein conformation in different experimental conditions, potentially requiring optimization of denaturation protocols for western blotting or fixation methods for immunohistochemistry. Given the high sequence homology between mouse, rat, and human HOXA11, many antibodies show cross-reactivity across these species, enabling comparative developmental studies .

What are the optimal storage and handling conditions for biotin-conjugated HOXA11 antibodies?

For maximum stability and performance, biotin-conjugated HOXA11 antibodies should be stored at -20°C in appropriate buffer conditions . The standard storage buffer typically consists of PBS (pH 7.3) containing 1% BSA, 50% glycerol, and 0.02% sodium azide to prevent microbial contamination and maintain antibody stability . When working with these antibodies, it's advisable to aliquot the stock solution to avoid repeated freeze-thaw cycles, which can degrade the antibody and reduce binding efficiency. For short-term storage (less than a week), antibodies can be kept at 4°C, but long-term storage should always be at -20°C. The stability period is typically 12 months from the date of receipt when stored properly . Due to the presence of biotin conjugation, these antibodies should be protected from light during handling to prevent photobleaching of the biotin molecule. Before use, allow the antibody to equilibrate to room temperature and gently mix by inversion rather than vortexing, which can damage the antibody structure and biotin conjugation.

How can researchers optimize immunofluorescence protocols when using biotin-conjugated HOXA11 antibodies?

Optimizing immunofluorescence protocols with biotin-conjugated HOXA11 antibodies requires careful attention to several experimental parameters. Begin with appropriate fixation - 4% paraformaldehyde is generally recommended for preserving protein structure while maintaining antigen accessibility . For intracellular targets like HOXA11, include a permeabilization step using 0.1-0.5% Triton X-100 in PBS for 10-15 minutes at room temperature. The critical blocking step should employ 5-10% normal serum (from the same species as the secondary reagent) with 1% BSA in PBS for 1 hour at room temperature to reduce non-specific binding.

When using the biotin-conjugated primary antibody, dilute to approximately 1:100 in blocking buffer and incubate overnight at 4°C in a humidified chamber . For detection, use fluorophore-conjugated streptavidin (e.g., streptavidin-Alexa Fluor) at 1:500-1:1000 dilution for 1 hour at room temperature, protected from light. Between all steps, perform multiple 5-minute washes with PBS containing 0.05% Tween-20. If tissues contain high endogenous biotin (common in kidney, liver, and brain), incorporate an avidin/biotin blocking step before primary antibody incubation. Finally, counterstain nuclei with DAPI and mount with an anti-fade mounting medium to preserve fluorescence signal during imaging and storage.

What strategies can resolve cross-reactivity issues when using HOXA11 antibodies in developmental studies?

Addressing cross-reactivity challenges in developmental studies using HOXA11 antibodies requires a multi-faceted approach. First, perform comprehensive antibody validation using positive and negative control tissues - embryonic tissues known to express HOXA11 versus those that don't, respectively . Implement peptide competition assays by pre-incubating the antibody with excess immunizing peptide (if available) to confirm binding specificity . The western blot should show band elimination when the antibody is pre-absorbed with the target peptide.

For tissues with potential cross-reactivity, optimize antibody dilution by testing a range (1:100 to 1:2000) to identify concentrations that maximize specific signal while minimizing background . Consider alternative fixation protocols, as overfixation can create artifacts and cross-reactivity. Employ thorough blocking with 5-10% normal serum plus 1% BSA, potentially adding 0.1-0.3% Triton X-100 to reduce non-specific hydrophobic interactions.

In multiplex studies, carefully sequence your staining protocol, completing HOXA11 detection before proceeding to other targets. For particularly challenging samples, consider antigen retrieval optimization by testing different buffers (citrate pH 6.0 versus EDTA pH 9.0) and retrieval times. Finally, validate results using orthogonal methods such as RNA in situ hybridization or RNAscope to confirm HOXA11 expression patterns independently of antibody-based detection.

How can biotin-conjugated HOXA11 antibodies be utilized in chromatin immunoprecipitation (ChIP) experiments?

Chromatin immunoprecipitation with biotin-conjugated HOXA11 antibodies offers significant advantages for studying transcription factor binding sites and epigenetic regulation. For optimal ChIP protocol implementation, begin with effective crosslinking using 1% formaldehyde for 10 minutes at room temperature, followed by quenching with 125 mM glycine. After cell lysis, sonicate chromatin to generate 200-500 bp fragments, which provides ideal resolution for transcription factor binding site identification.

For the immunoprecipitation step, pre-clear chromatin with protein A/G beads for 1 hour at 4°C to reduce non-specific binding. Incubate cleared chromatin with biotin-conjugated HOXA11 antibody (2-5 μg per reaction) overnight at 4°C with gentle rotation . Rather than using protein A/G beads directly, capture the biotin-conjugated antibody-chromatin complexes with streptavidin-coated magnetic beads, which provides higher affinity and cleaner pull-down compared to traditional approaches. After capture, perform stringent washing steps with increasing salt concentrations to remove non-specific interactions.

For elution, reverse crosslinks by incubating at 65°C for 4-6 hours, followed by proteinase K and RNase A treatment. The purified DNA can then be analyzed by qPCR for known targets or subjected to next-generation sequencing (ChIP-seq) for genome-wide binding site identification. For ChIP-seq applications, include input controls and IgG controls to facilitate peak calling and identification of true binding events. This approach enables comprehensive mapping of HOXA11 binding sites in developmental contexts or reproductive tissues.

What approaches can enhance detection sensitivity when working with low HOXA11 expression levels?

Enhancing detection sensitivity for low-abundance HOXA11 requires systematic optimization of multiple experimental parameters. Begin by selecting the appropriate biotin-conjugated HOXA11 antibody with proven sensitivity for your application and species of interest . For protein detection methods, concentrate samples using immunoprecipitation before western blotting to enrich for HOXA11. Utilize enhanced chemiluminescence (ECL) substrates with extended signal duration for western blotting, and consider tyramide signal amplification (TSA) for immunohistochemistry and immunofluorescence applications.

When working with biotin-conjugated antibodies specifically, leverage the strong biotin-streptavidin interaction (Kd ≈ 10^-15 M) by using streptavidin conjugated to high-quantum-yield fluorophores or enzymes with high turnover rates . For immunofluorescence, employ confocal microscopy with photomultiplier tube detectors set to higher sensitivity, or consider super-resolution techniques for localization studies. Reduce background by extending blocking times (2-3 hours) with 5% normal serum and 1% BSA, and include 0.1-0.3% Triton X-100 to minimize non-specific hydrophobic interactions.

For ELISA applications, implement sandwich ELISA formats with a capture antibody against HOXA11 and detection using the biotin-conjugated antibody, followed by streptavidin-HRP. Signal development with fluorogenic or chemiluminescent substrates provides greater sensitivity than chromogenic detection. Finally, for quantitative applications, construct standard curves using recombinant HOXA11 protein to establish the lower limit of detection and ensure measurements fall within the linear dynamic range of the assay.

How can HOXA11 antibodies be utilized to investigate interactions with other transcription factors?

Investigating HOXA11 interactions with other transcription factors requires specialized approaches leveraging biotin-conjugated antibodies. For co-immunoprecipitation studies, cell or tissue lysates should be prepared using gentle lysis buffers (containing 150 mM NaCl, 1% NP-40 or 0.5% Triton X-100, 50 mM Tris pH 8.0) supplemented with protease inhibitors and phosphatase inhibitors to preserve protein complexes . Pre-clear lysates with streptavidin beads before incubating with biotin-conjugated HOXA11 antibody overnight at 4°C. After capturing complexes with fresh streptavidin beads, wash stringently but gently to maintain protein-protein interactions.

For visualizing co-localization, proximity ligation assay (PLA) offers superior resolution compared to standard co-immunofluorescence. In this technique, use biotin-conjugated HOXA11 antibody with a primary antibody against the potential interacting partner, followed by appropriate secondary antibodies conjugated to PLA probes. When the proteins are in close proximity (<40 nm), the PLA probes enable ligation and rolling circle amplification, generating a fluorescent spot visible by microscopy.

Chromatin immunoprecipitation followed by sequential ChIP (ChIP-reChIP) can identify genomic regions co-occupied by HOXA11 and other factors. After performing initial ChIP with biotin-conjugated HOXA11 antibody and streptavidin beads, elute complexes under mild conditions, then perform a second ChIP with antibodies against suspected interacting partners. This approach reveals genomic loci where both factors are simultaneously bound. Complementary approaches include bioluminescence resonance energy transfer (BRET) or fluorescence resonance energy transfer (FRET) with appropriately tagged proteins to study interactions in living cells.

How can researchers validate the specificity of biotin-conjugated HOXA11 antibodies?

Comprehensive validation of biotin-conjugated HOXA11 antibodies requires a multi-technique approach to ensure experimental reliability. Begin with western blot analysis using positive control lysates from tissues or cell lines known to express HOXA11 (such as developing limb buds or reproductive tissues) alongside negative controls . Verify that the observed band appears at the expected molecular weight (approximately 34 kDa, though it may appear around 30 kDa) . Perform peptide competition assays by pre-incubating the antibody with excess immunizing peptide, which should eliminate specific bands on western blot and signals in immunostaining if the antibody is truly specific .

For genetic validation, compare antibody staining in wild-type versus HOXA11 knockout or knockdown models when available. Alternatively, overexpress tagged HOXA11 in a cell line with low endogenous expression and confirm co-localization of antibody signal with the tag. RNA-protein correlation studies using RT-qPCR or RNA-seq data compared to protein detection can provide additional validation, as protein and mRNA levels should generally correlate in different tissues or developmental stages.

For biotin conjugation quality, confirm functional conjugation by performing a simple ELISA or dot blot using streptavidin-HRP directly against the antibody. To exclude potential cross-reactivity with other HOX family members, test the antibody against recombinant HOXA10, HOXA13, and other closely related HOX proteins. Finally, check lot-to-lot consistency by maintaining reference samples and comparing results across different antibody lots to ensure reproducibility in your experimental system.

What are common pitfalls when using biotin-conjugated antibodies in tissues with high endogenous biotin?

Working with biotin-conjugated antibodies in tissues with high endogenous biotin presents several challenges that require specific methodological adaptations. Tissues rich in endogenous biotin include liver, kidney, brain, and adipose tissue, which can generate significant background signal. The fundamental approach to addressing this issue is implementing an avidin/biotin blocking step before applying the biotin-conjugated primary antibody .

A comprehensive blocking protocol involves first treating tissue sections with avidin solution (100-500 μg/ml in PBS) for 15-20 minutes, which binds to endogenous biotin. After a brief rinse, apply biotin solution (100-500 μg/ml in PBS) for another 15-20 minutes to saturate any remaining avidin binding sites. This two-step process effectively blocks endogenous biotin without interfering with subsequent detection steps.

Additionally, consider alternative fixation protocols, as some fixatives can enhance biotin accessibility. Paraformaldehyde fixation (2-4%) is often preferable to harsher fixatives like Bouin's solution. If background persists, implement more stringent blocking with 0.1% streptavidin and 0.01% biotin in blocking buffer for 30 minutes each.

For critical experiments, consider alternative detection strategies such as non-biotin polymeric detection systems or directly conjugated fluorophores. Always include appropriate controls: (1) a section with no primary antibody to assess streptavidin background, (2) a section without streptavidin detection to evaluate autofluorescence, and (3) a competitive blocking control with excess free biotin. These approaches ensure reliable results even in tissues with challenging endogenous biotin levels.

How can researchers quantitatively analyze HOXA11 expression levels using biotin-conjugated antibodies?

Quantitative analysis of HOXA11 expression using biotin-conjugated antibodies requires rigorous standardization and appropriate controls. For western blot quantification, include a standard curve using recombinant HOXA11 protein at known concentrations (typically 5-100 ng) alongside your samples . Ensure equal loading using housekeeping proteins like β-actin or GAPDH, and capture images within the linear dynamic range of your detection system. For densitometric analysis, use software like ImageJ to measure band intensity, normalizing HOXA11 signals to loading controls.

For immunohistochemistry quantification, implement systematic unbiased sampling techniques such as the optical fractionator or dissector methods. Standardize all staining parameters including antibody concentration (typically 1:150 for IHC applications), incubation times, and development conditions . Use automated image analysis software with appropriate thresholding to quantify staining intensity or positive cell counts, ensuring consistency across all samples.

Flow cytometry offers powerful quantitative capabilities for cell-by-cell analysis. Establish a standardized staining protocol using biotin-conjugated HOXA11 antibody at 1:100 dilution followed by fluorophore-conjugated streptavidin . Include fluorescence minus one (FMO) controls and isotype controls to set accurate gates. For absolute quantification, use calibration beads with known quantities of fluorophore to convert fluorescence intensity to molecules of equivalent soluble fluorochrome (MESF).

For all quantitative applications, implement appropriate statistical analyses based on sample size and distribution characteristics. Report central tendency (mean or median) along with dispersion measures (standard deviation or interquartile range) and perform appropriate significance testing (t-tests, ANOVA, or non-parametric alternatives) when comparing groups. This comprehensive approach ensures scientifically valid quantitative assessment of HOXA11 expression across experimental conditions.

How can biotin-conjugated HOXA11 antibodies be utilized in single-cell protein analysis techniques?

Single-cell protein analysis with biotin-conjugated HOXA11 antibodies represents a frontier in developmental biology research. Mass cytometry (CyTOF) offers a powerful approach by leveraging the biotin-streptavidin interaction coupled with metal-tagged streptavidin. For optimal implementation, fix cells with 1.6% paraformaldehyde for 10 minutes at room temperature, then permeabilize with 0.1% saponin in cell staining buffer. Block with 10% serum and 1% BSA before adding biotin-conjugated HOXA11 antibody at 1:50 dilution . After washing, detect with heavy metal-conjugated streptavidin (typically lanthanide metals). This approach enables simultaneous analysis of HOXA11 along with dozens of other proteins without fluorescence spectral overlap concerns.

For imaging-based single-cell analysis, implement imaging mass cytometry or multiplexed ion beam imaging (MIBI), which utilize similar metal-tagging strategies but preserve spatial information. Alternatively, cyclic immunofluorescence (CycIF) allows iterative staining, imaging, and signal removal, enabling visualization of dozens of proteins including HOXA11 in the same tissue section. For each cycle, use biotin-conjugated HOXA11 antibody followed by fluorophore-conjugated streptavidin, image, then chemically quench or remove the signal before the next staining round.

Single-cell western blotting platforms can separate proteins from individual cells on specialized gels, followed by transfer and detection with biotin-conjugated HOXA11 antibody and streptavidin-HRP. For microfluidic approaches, capture cells in droplets or microwells, then perform proximity assays with biotin-conjugated HOXA11 antibody paired with DNA-barcoded streptavidin for subsequent sequencing readout. These innovative approaches reveal cell-to-cell heterogeneity in HOXA11 expression within developing tissues or disease states, providing insights inaccessible through bulk analysis methods.

What considerations are important when designing multiplex experiments using biotin-conjugated HOXA11 antibodies?

Designing effective multiplex experiments with biotin-conjugated HOXA11 antibodies requires careful planning to avoid interference between detection systems. First, consider the detection strategy—since biotin-conjugated antibodies require streptavidin detection, ensure other primary antibodies in your panel use different detection systems, such as directly conjugated fluorophores or species-specific secondary antibodies .

For optimal panel design, select primary antibodies raised in different host species to avoid cross-reactivity. When this isn't possible, implement sequential staining protocols with complete blocking steps between detection cycles. Start with the lowest abundance target (often HOXA11 in non-developmental tissues) to maximize detection sensitivity before proceeding to more abundant proteins. If using tyramide signal amplification (TSA) with HRP-streptavidin, perform this step first, followed by heat-mediated antibody stripping (70°C in citrate buffer for 20 minutes) before subsequent staining rounds.

To minimize bleed-through in fluorescence applications, choose fluorophores with well-separated excitation and emission spectra. For each fluorophore combination, prepare single-stained controls to establish compensation settings for flow cytometry or confocal microscopy. Include an unstained control and fluorescence minus one (FMO) controls for accurate gating and background assessment. When multiplex imaging is performed, include a reference channel (typically DAPI for nuclei) in all acquisition rounds to enable precise image registration during analysis.

For quality control, validate each antibody individually before combining them, ensuring specific staining patterns and expected cellular localization. Finally, prepare a detailed experimental template documenting all antibody concentrations, incubation times, and washing steps to ensure reproducibility across experiments. This systematic approach maximizes information yield while maintaining data integrity in complex multiplex studies.

How can researchers leverage biotin-conjugated HOXA11 antibodies for spatial transcriptomics studies?

Integrating biotin-conjugated HOXA11 antibodies with spatial transcriptomics creates powerful opportunities for correlating protein expression with transcriptional profiles in a spatial context. For Visium (10x Genomics) or similar spatial transcriptomics platforms, perform sequential workflow optimization beginning with RNA capture and sequencing according to standard protocols. Subsequently on consecutive sections, conduct immunohistochemistry using biotin-conjugated HOXA11 antibody (1:150 dilution) followed by streptavidin-HRP and chromogenic detection . Digitize both the transcriptomics results and immunostained sections, then align images using morphological landmarks or reference stains.

For higher-resolution co-detection approaches, implement advanced techniques like Immuno-SABER (Signal Amplification By Exchange Reaction), which combines protein detection with transcriptome analysis. Apply biotin-conjugated HOXA11 antibody, followed by DNA-barcoded streptavidin conjugates carrying unique sequence identifiers. These DNA barcodes can be amplified through programmed hybridization chain reactions, generating substantial signal amplification while maintaining spatial resolution.

Alternatively, for a more integrated approach, use CITE-seq principles in a spatial context by applying DNA-barcoded streptavidin to sections after biotin-conjugated HOXA11 antibody binding. The DNA barcodes are later sequenced alongside spatially-resolved transcriptomics data, providing direct correlation between HOXA11 protein presence and gene expression profiles.

For data analysis, implement computational workflows that integrate protein expression intensity with transcriptomic clusters. Correlation analyses between HOXA11 protein levels and mRNA expression of known target genes can reveal post-transcriptional regulation mechanisms. Dimensionality reduction techniques like UMAP or t-SNE performed on integrated protein-RNA datasets help identify spatial domains with unique molecular signatures. This integrated approach bridges protein localization with comprehensive gene expression profiles while preserving critical spatial information in developmental contexts.

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.