HORMAD1 Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to HORMAD1 Antibody

HORMAD1 (HORMA Domain-Containing Protein 1) antibodies are immunological tools designed to detect and study the HORMAD1 protein, a meiosis-specific protein aberrantly expressed in various cancers. These antibodies enable researchers to investigate HORMAD1's roles in DNA repair, homologous recombination (HR), and cancer biology, particularly in contexts such as chemotherapy resistance and genomic instability .

Recommended Dilutions and Protocols

  • Western Blot (WB): 1:500–1:2000 dilution .

  • Immunofluorescence (IF): Optimized for detecting nuclear HORMAD1 foci in cancer cells .

  • Immunohistochemistry (IHC): Validated for paraffin-embedded tumor samples .

Physiological Role in Meiosis

HORMAD1 regulates homologous chromosome pairing and crossover formation during meiosis via its HORMA domain, ensuring proper synaptonemal complex assembly . It dynamically localizes to chromosomal axes during prophase I and dissociates post-synapsis .

Oncogenic Roles in Cancer

HORMAD1 is overexpressed in lung adenocarcinoma (LUAD), ovarian carcinoma, and triple-negative breast cancer (TNBC), where it promotes:

  • Chemoresistance: Enhances survival under genotoxic stress (e.g., docetaxel, cisplatin) .

  • DNA Repair: Facilitates HR-mediated repair by promoting RAD51 filament formation and resection of DNA double-strand breaks (DSBs) .

  • Angiogenesis: Silencing HORMAD1 reduces VEGF levels and microvessel density in ovarian cancer xenografts .

DNA Damage Response Mechanisms

  • HR Promotion: HORMAD1 depletion reduces RAD51 foci formation and sensitizes cells to γ-irradiation and PARP inhibitors .

  • Resection Dependency: HORMAD1 enables ssDNA formation at DSBs, a critical step for HR .

  • Germline vs. Cancer Roles: Unlike its meiosis-specific DSB suppression in germ cells, HORMAD1 adopts a pro-repair role in cancer cells .

Genomic Instability and Mismatch Repair

HORMAD1 disrupts nuclear localization of the MCM8–MCM9 complex, impairing MLH1 chromatin binding and mismatch repair (MMR). This correlates with increased mutation load in HORMAD1-expressing tumors .

Prognostic and Predictive Biomarker

  • Poor Prognosis: High HORMAD1 expression associates with advanced tumor stage and resistance to platinum-based therapies in LUAD and TNBC .

  • Therapeutic Target: siRNA-mediated HORMAD1 silencing synergizes with cisplatin, reducing tumor weight and ascites in ovarian cancer models .

Immunotherapeutic Potential

As a cancer/testis antigen, HORMAD1’s aberrant expression in tumors makes it a candidate for vaccine development or CAR-T cell therapy .

Antibody Validation Data

  • Western Blot: Detects endogenous HORMAD1 at ~45–55 kDa in HUVEC cells and cancer lines .

  • Immunofluorescence: Localizes to nuclear foci co-staining with γH2AX post-irradiation .

  • Knockdown/Knockout Controls: Reduced signal in siRNA-treated cells confirms specificity .

Limitations

  • Isoform Variability: Observed molecular weights vary due to post-translational modifications .

  • Context-Dependent Effects: HORMAD1’s role in HR remains controversial in certain cancers (e.g., conflicting reports in TNBC) .

Product Specs

Buffer
Phosphate Buffered Saline (PBS) with 0.1% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid repeated freeze-thaw cycles.
Lead Time
We typically dispatch products within 1-3 business days of receiving your order. Delivery times may vary depending on the shipping method and destination. For specific delivery time estimates, please contact your local distributor.
Synonyms
HORMAD1 antibody; NOHMA antibody; HORMA domain-containing protein 1 antibody; Cancer/testis antigen 46 antibody; CT46 antibody; Newborn ovary HORMA protein antibody
Target Names
HORMAD1
Uniprot No.

Target Background

Function

HORMAD1 Antibody plays a pivotal role in meiotic progression. It regulates three distinct functions during meiosis:

  • **Enhances DSB processing:** HORMAD1 ensures an adequate number of processed DNA double-strand breaks (DSBs) are available for successful homologous recombination by increasing the steady-state levels of single-stranded DSB ends.
  • **Promotes synaptonemal complex formation:** HORMAD1 facilitates the formation of the synaptonemal complex independently of its role in homology search.
  • **Meiotic checkpoint control:** HORMAD1 plays a crucial role in the male mid-pachytene checkpoint and the female meiotic prophase checkpoint. It is essential for the efficient accumulation of ATR activity on unsynapsed chromosome regions, a process believed to underlie meiotic silencing of unsynapsed chromatin (MSUC) and meiotic prophase quality control in both sexes.
Gene References Into Functions
  1. Genomic Complexity Profiling Reveals That HORMAD1 Overexpression Contributes to Homologous Recombination Deficiency in Triple-Negative Breast Cancers. PMID: 25770156
  2. Hormad1 and Hormad2 may not be the susceptible genes for non-obstructive azoospermia in the study population. However, rs718772 of Hormad2 variant might be associated with testis development in NOA patients. PMID: 24803422
  3. HORMAD1 gene silencing resulted in significantly reduced VEGF protein levels and microvessel density compared to controls in epithelial ovarian carcinoma. PMID: 22776561
  4. This study is the first to demonstrate that HORMAD1 single-nucleotide polymorphisms (SNPs) may predispose men to a defect in spermatogenesis. However, the causal and potential mechanistic relationships between these HORMAD1 SNPs and azoospermia remain unclear. PMID: 22407170
Database Links

HGNC: 25245

OMIM: 609824

KEGG: hsa:84072

STRING: 9606.ENSP00000355167

UniGene: Hs.298312

Subcellular Location
Nucleus. Chromosome.
Tissue Specificity
Testis-specific. Over-expressed in carcinomas.

Q&A

What is HORMAD1 and what are its primary biological functions?

HORMAD1 (also known as NOHMA, Cancer/testis antigen 46, Newborn ovary HORMA protein, or CT46) is a meiosis-specific protein that regulates three critical functions during meiosis. First, it ensures sufficient processed DNA double-strand breaks (DSBs) are available for successful homology search by increasing single-stranded DSB ends. Second, it promotes synaptonemal complex formation independently of its homology search role. Third, it plays a crucial role in the male mid-pachytene checkpoint and female meiotic prophase checkpoint . HORMAD1 is required for efficient ATR activity build-up on unsynapsed chromosome regions, forming the basis of meiotic silencing of unsynapsed chromatin (MSUC) and quality control mechanisms during meiotic prophase in both sexes .

How do I select the appropriate HORMAD1 antibody for my experimental design?

Selection of an appropriate HORMAD1 antibody depends on several experimental factors:

  • Application compatibility: Verify the antibody is validated for your specific application (ICC/IF, ELISA, Western blot, etc.). For example, ab220021 is suitable for ICC/IF with human samples .

  • Host species compatibility: Consider the host species (e.g., rabbit polyclonal) in relation to your secondary detection system and other antibodies in multiplexed experiments.

  • Epitope recognition: Check the immunogen details (e.g., recombinant fragment within Human HORMAD1 aa 250-350) or (recombinant human HORMA domain-containing protein 1 protein 201-394AA) to ensure the antibody recognizes relevant domains for your study.

  • Validated reactivity: Confirm the antibody has been tested in your species of interest, as cross-reactivity varies between antibodies.

  • Conjugation requirements: Determine if you need unconjugated or conjugated (e.g., HRP-conjugated) antibodies based on your detection system .

What are the recommended sample preparation methods for HORMAD1 immunodetection?

For optimal HORMAD1 immunodetection in different experimental contexts:

Sample TypeFixation MethodRecommended ProtocolNotes
Cell culturesPFA-fixation with Triton X-100 permeabilization4% PFA for 10-15 minutes followed by 0.1-0.5% Triton X-100Preserves protein localization while allowing antibody access
Tissue sectionsParaffin-embedded or frozen sectionsStandard antigen retrieval methodsMay require optimization for specific tissues
Chromosome spreads1% PFA with 0.15% Triton X-100Specialized spreading techniquesCritical for meiotic studies
Protein extractsN/AStandard protein extraction buffers with protease inhibitorsUse fresh samples when possible

For chromosome spread assays specifically, the methodology used in Hormad1 knockout studies involves: antibody detection with anti-SYCP2 combined with markers like DMC1, RAD51, RPA, and γH2AX to visualize components of the meiotic recombination machinery .

How can I optimize co-immunostaining of HORMAD1 with other meiotic proteins?

Optimizing co-immunostaining of HORMAD1 with other meiotic proteins requires careful consideration of antibody compatibility and sample preparation:

  • Sequential antibody application: When using multiple primary antibodies from the same host species, apply them sequentially with blocking steps in between. For example, when examining HORMAD1 with recombination proteins like RAD51 or DMC1, apply primary antibodies in order of sensitivity.

  • Fluorophore selection: Choose fluorophores with minimal spectral overlap (e.g., Alexa Fluor 488 for SYCP2 and Alexa Fluor 555 for HORMAD1) to avoid bleed-through in microscopy .

  • Validation controls: Always include single-stained controls and minus-primary controls to verify antibody specificity and prevent misinterpretation of co-localization signals.

  • Sample preparation optimization: For chromosome spreads, the 1% PFA fixation protocol with 0.15% Triton X-100 permeabilization has been effectively used in studies comparing wild-type and Hormad1−/− tissues .

  • Signal amplification: For weak signals, consider using biotinylated secondary antibodies followed by streptavidin-conjugated fluorophores or tyramide signal amplification.

The synaptonemal complex protein SYCP2 serves as an excellent marker for chromosome axes and can be effectively co-stained with HORMAD1 .

How does HORMAD1 antibody staining pattern differ between normal meiotic cells and cancer cells with aberrant HORMAD1 expression?

The staining patterns of HORMAD1 differ significantly between normal meiotic cells and cancer cells:

Normal meiotic cells:

  • Linear staining along unsynapsed chromosome axes during leptotene and zygotene stages

  • Disappearance from synapsed regions during pachytene

  • Retention on unsynapsed regions of sex chromosomes in males

  • Association with ATR, BRCA1, and γH2AX on unsynapsed chromosomes

Cancer cells with aberrant HORMAD1 expression:

  • Predominantly nuclear localization in cancer cells

  • Association with chromatin and the replisome in lung adenocarcinoma cells

  • Co-localization with replication fork components and protection factors like RAD51

  • Punctate nuclear pattern during S-phase, particularly at sites of replication stress

This differential staining pattern provides important insights for researchers investigating the pathological roles of HORMAD1 in cancers versus its normal meiotic functions. In particular, HORMAD1's association with RAD51 and the replisome in cancer cells suggests a role in protecting stalled replication forks from degradation - a function distinct from its meiotic role .

What are the recommended protocols for quantifying HORMAD1 localization and abundance in experimental samples?

For rigorous quantification of HORMAD1 localization and abundance:

  • Immunofluorescence intensity quantification:

    • Capture images using identical microscope settings for all samples

    • Use software like ImageJ/FIJI to measure integrated density or mean fluorescence intensity

    • Compare signal intensity between wild-type and experimental conditions (e.g., 76% decrease in γH2AX signal intensity in Hormad1−/− spermatocytes)

  • Western blot quantification:

    • Use standardized loading controls (β-actin, GAPDH, or Histone-H3)

    • Perform densitometry analysis with normalization to housekeeping proteins

    • Include a standard curve if absolute quantification is required

  • Chromatin association analysis:

    • Perform cellular fractionation to separate chromatin-bound from soluble proteins

    • Compare HORMAD1 levels in different cellular compartments

    • Use histone markers (e.g., Histone-H3) to verify chromatin fraction purity

  • Co-localization analysis:

    • Calculate Pearson's or Mander's coefficients for co-localization with proteins of interest

    • Use specialized co-localization plugins in ImageJ or other image analysis software

    • Perform statistical analysis comparing co-localization in different experimental conditions

How does HORMAD1 affect homologous recombination in cancer cells versus meiotic cells?

HORMAD1 has distinct and sometimes opposing effects on homologous recombination (HR) in cancer cells versus meiotic cells:

In meiotic cells:

  • Ensures sufficient DSB formation for homologous chromosome pairing

  • Promotes interhomolog recombination over sister chromatid repair

  • Required for efficient RAD51/DMC1 loading at DSB sites

  • Essential for proper synaptonemal complex formation

In cancer cells:

  • Context-dependent effects varying by cancer type:

    In triple-negative breast cancer:

    • Suppresses conservative HR (55% reduction in HR in SUM159 cells overexpressing HORMAD1)

    • Increases non-homologous end joining (NHEJ) activity

    • Increases sensitivity to PARP inhibitors like olaparib and BMN673

    • Promotes genomic instability

    In lung adenocarcinoma:

    • Protects stalled replication forks from MRE11-mediated degradation

    • Stabilizes RAD51 on chromatin

    • Prevents excessive accumulation of DNA damage

    • Protects against genomic instability

These opposing roles of HORMAD1 in different cancer contexts highlight the importance of cancer-type specific investigation. The shift from promoting genomic instability in breast cancer to protecting against it in lung adenocarcinoma represents a fascinating area for further research into context-dependent functions .

What experimental approaches can detect HORMAD1-dependent alterations in DNA repair pathways?

Several experimental approaches effectively measure HORMAD1's impact on DNA repair:

  • DR-GFP assay for HR efficiency:

    • Utilizes I-SceI endonuclease-induced double-strand breaks

    • Detected 55% reduction in HR in SUM159 cells overexpressing HORMAD1

    • Detected 36% decrease in HR activity in CAL51 cells overexpressing HORMAD1

    • Showed 100% and 55% increases in HR following HORMAD1 knockdown in HCC1143 and MDA-MB-436 cells, respectively

  • EJ5 NHEJ GFP reporter assay:

    • Measures non-homologous end joining activity

    • Demonstrated increased NHEJ activity in HORMAD1-overexpressing cells

    • Showed reduced NHEJ activity following HORMAD1 knockdown in HCC1143 cells

  • 53BP1 focus formation assay:

    • Quantifies DNA repair focus formation following irradiation or hydroxyurea treatment

    • HORMAD1 overexpression increased 53BP1 focus formation

    • HORMAD1 knockdown reduced damage-induced 53BP1 focus formation

  • Replication fork protection assays:

    • DNA fiber analysis to measure replication fork degradation

    • Demonstrates HORMAD1's role in protecting stalled replication forks from MRE11-mediated degradation in lung adenocarcinoma

  • Drug sensitivity assays:

    • Tests for synthetic lethality with PARP inhibitors (olaparib, BMN673)

    • Cisplatin sensitivity assessments

    • HORMAD1 overexpression increases sensitivity to PARP inhibitors in breast cancer models

What are the technical challenges in differentiating HORMAD1-specific effects from other DNA repair alterations in experimental models?

Researchers face several technical challenges when isolating HORMAD1-specific effects:

  • Redundancy in DNA repair pathways: HORMAD1 influences multiple repair mechanisms, making it difficult to attribute phenotypes to specific pathways. Control experiments should include markers for various repair pathways (HR, NHEJ, MMEJ).

  • Cancer heterogeneity: As seen in the contrasting roles in breast cancer versus lung adenocarcinoma, HORMAD1's function is context-dependent. Researchers should characterize baseline repair defects in their specific model systems .

  • Long-term adaptation: Long-term HORMAD1 expression can trigger compensatory mechanisms. For example, while HORMAD1 expression induced cisplatin sensitivity, knockdown in long-term expressing cells didn't always reverse this phenotype, suggesting adaptation mechanisms .

  • Pleiotropic effects: HORMAD1 influences both meiotic recombination and replication stress responses. Studies need to carefully control for cell cycle phase and differentiate between these distinct functions.

  • Technical variability in antibody specificity: Different antibodies may recognize distinct HORMAD1 conformations or complexes. Validation with multiple antibodies and genetic models (siRNA, CRISPR) is essential.

To address these challenges, researchers should employ genetic rescue experiments, domain-specific mutations, and comprehensive pathway analysis rather than relying on single assays or models.

HORMAD1 in Cancer Research

Researchers can employ several methodologies to assess HORMAD1's impact on genomic stability:

  • Allelic imbalance profiling:

    • Whole-genome sequencing or SNP arrays to detect copy number variations

    • Analysis of large-scale state transitions (LSTs)

    • Measurement of telomeric allelic imbalance (TAI)

    • Detection of loss of heterozygosity (LOH) patterns

  • Chromosomal instability assays:

    • Metaphase spread analysis for structural chromosomal aberrations

    • Micronuclei formation quantification

    • Anaphase bridge detection

    • Fluorescence in situ hybridization (FISH) for specific chromosomal rearrangements

  • DNA damage marker quantification:

    • Immunofluorescence quantification of γH2AX foci (76% decrease in signal intensity in Hormad1−/− spermatocytes)

    • 53BP1 focus formation analysis

    • RAD51 focus quantification

  • Mutational signature analysis:

    • Whole-genome sequencing to identify characteristic mutational patterns

    • Assessment of specific mutation types (transitions, transversions, indels)

    • Correlation of mutation burden with HORMAD1 expression levels

  • Single-cell genomics:

    • Single-cell DNA sequencing to assess heterogeneity in genomic instability

    • Correlation of copy number aberrations with HORMAD1 expression at single-cell resolution

These methodologies together provide a comprehensive assessment of how HORMAD1 influences genomic stability, important for understanding its role in cancer progression and therapeutic response.

How can researchers distinguish between correlation and causation in studies of HORMAD1's role in cancer progression?

Distinguishing correlation from causation in HORMAD1 cancer research requires rigorous experimental approaches:

  • Genetic manipulation experiments:

    • Knockdown/knockout studies using siRNA or CRISPR-Cas9 (as demonstrated in HCC1143 and MDA-MB-436 cells)

    • Overexpression studies with appropriate controls (as shown in SUM159-HORMAD1-V5 versus SUM159-LacZ-V5 cells)

    • Rescue experiments with wild-type versus mutant HORMAD1

  • Time-course analyses:

    • Inducible expression systems to monitor temporal changes following HORMAD1 activation

    • Assess immediate versus long-term consequences of HORMAD1 expression

    • Document adaptation mechanisms (as observed in long-term HORMAD1-expressing cells)

  • Domain-specific mutations:

    • Create variants with specific domain disruptions to map functional requirements

    • Test HORMAD domain versus other protein regions for specific phenotypes

    • Identify separation-of-function mutations that affect cancer but not meiotic functions

  • Clinical sample validation:

    • Correlate in vitro findings with patient sample analyses

    • Stratify patient cohorts by HORMAD1 expression levels

    • Compare treatment responses in HORMAD1-high versus HORMAD1-low tumors

  • Mechanistic pathway dissection:

    • Use epistasis experiments with other DNA repair factors

    • Perform direct biochemical assays for HORMAD1 function

    • Identify direct interaction partners in cancer versus meiotic contexts

The study by Watkins et al. provides an exemplary approach by demonstrating both correlation (HORMAD1 expression associated with specific tumor types) and causation (direct effects of HORMAD1 manipulation on HR, NHEJ, and drug sensitivity) .

What are common causes of inconsistent HORMAD1 antibody staining and how can they be addressed?

Researchers often encounter several challenges with HORMAD1 antibody staining:

  • Fixation artifacts:

    • Problem: Overfixation can mask epitopes while underfixation leads to poor morphology

    • Solution: Optimize fixation time and concentration (standard is 4% PFA for 10-15 minutes)

    • Validation: Compare multiple fixation protocols with positive controls

  • Antibody specificity issues:

    • Problem: Cross-reactivity with related HORMA domain proteins

    • Solution: Validate antibodies using Hormad1 knockout tissues as negative controls

    • Validation: Perform peptide competition assays to confirm specificity

  • Cell cycle-dependent expression:

    • Problem: HORMAD1 expression and localization vary across meiotic stages

    • Solution: Use stage-specific markers (SYCP2, SYCP3) to properly identify cell cycle phase

    • Validation: Compare with established stage-specific expression patterns

  • Signal-to-noise ratio:

    • Problem: High background obscuring specific HORMAD1 signal

    • Solution: Optimize blocking conditions (BSA, normal serum from secondary antibody host)

    • Validation: Include secondary-only controls to assess background

  • Protein complex masking epitopes:

    • Problem: HORMAD1 in protein complexes may have masked epitopes

    • Solution: Test multiple antibodies targeting different epitopes

    • Validation: Compare staining patterns between antibodies recognizing different regions

How can researchers validate HORMAD1 antibody specificity in their experimental systems?

Thorough validation of HORMAD1 antibodies is critical for experimental reliability:

  • Genetic validation:

    • Use tissues/cells from Hormad1 knockout models as negative controls

    • Apply HORMAD1 siRNA knockdown in positive cell lines to confirm specificity

    • Perform rescue experiments with HORMAD1 overexpression in knockout backgrounds

  • Multi-antibody comparison:

    • Compare staining patterns using antibodies targeting different HORMAD1 epitopes

    • Verify consistent localization patterns between different antibodies

    • Document any epitope-specific differences in accessibility

  • Western blot validation:

    • Confirm single band of expected molecular weight (approximately 41 kDa)

    • Verify band disappearance in knockout or knockdown samples

    • Test multiple tissues to confirm expression in expected cell types only

  • Peptide competition assays:

    • Pre-incubate antibody with immunizing peptide before application

    • Verify signal disappearance when the specific epitope is blocked

    • Include control peptides to confirm specificity of competition

  • Immunoprecipitation-mass spectrometry:

    • Perform IP with HORMAD1 antibody followed by mass spectrometry

    • Confirm HORMAD1 as the predominant precipitated protein

    • Identify any cross-reactive proteins for awareness in interpretation

How should researchers address contradictory findings between different HORMAD1 antibodies or detection methods?

When faced with contradictory results, researchers should:

  • Systematic comparison:

    • Create a table documenting specific differences between antibodies:

      • Epitope location

      • Host species

      • Validation methods used

      • Buffer and fixation conditions

    • Test all antibodies under identical conditions

  • Methodological triangulation:

    • Employ multiple detection techniques (immunofluorescence, Western blot, flow cytometry)

    • Compare protein-level results with mRNA expression data

    • Use proximity ligation assays to verify protein interactions in situ

  • Context-dependent analysis:

    • Assess if contradictions relate to specific cell types or experimental conditions

    • Consider the possibility of post-translational modifications affecting epitope recognition

    • Evaluate if protein complex formation might mask certain epitopes

  • Independent verification:

    • Use genetic approaches (CRISPR knockout, siRNA) to confirm antibody specificity

    • Apply techniques that don't rely on antibodies (e.g., fluorescent protein tagging)

    • Consult published literature for similar discrepancies and their resolutions

  • Reporting transparency:

    • Document all contradictory findings in publications

    • Provide complete antibody information (catalog numbers, lot numbers)

    • Share validation data even when results are inconsistent

The contradictory findings about HORMAD1's role in genomic stability between breast cancer and lung adenocarcinoma studies illustrate how context-dependent functions may be misinterpreted as contradictory results .

What are emerging methods for studying HORMAD1's interactions with the DNA repair machinery?

Cutting-edge approaches for investigating HORMAD1 interactions include:

  • Proximity-based labeling techniques:

    • BioID or TurboID fusion proteins to identify proximal interaction partners

    • APEX2-based proximity labeling for temporal mapping of HORMAD1 interactions

    • Split-BioID for detecting specific protein-protein interactions in living cells

  • Live-cell imaging of DNA repair dynamics:

    • CRISPR-mediated endogenous tagging of HORMAD1 with fluorescent proteins

    • Fluorescence recovery after photobleaching (FRAP) to assess protein dynamics

    • Single-molecule tracking to monitor HORMAD1 behavior at DNA damage sites

  • Chromatin immunoprecipitation sequencing (ChIP-seq):

    • Genome-wide mapping of HORMAD1 binding sites

    • Integration with DNA damage markers (γH2AX ChIP-seq)

    • Correlation with replication stress regions or common fragile sites

  • Cryo-electron microscopy:

    • Structural analysis of HORMAD1 in complex with DNA repair proteins

    • Visualization of HORMAD1 at replication forks

    • Conformational changes associated with DNA binding

  • Proteomics approaches:

    • Quantitative interaction proteomics before and after DNA damage

    • Post-translational modification mapping under different conditions

    • Crosslinking mass spectrometry to identify direct binding interfaces

These emerging technologies will help resolve the seemingly contradictory roles of HORMAD1 in different cancer contexts by providing higher-resolution data on its molecular interactions and functions.

How might single-cell approaches enhance our understanding of HORMAD1 function in heterogeneous cancer populations?

Single-cell methodologies offer unprecedented insights into HORMAD1 biology:

  • Single-cell RNA sequencing:

    • Correlation of HORMAD1 expression with DNA repair gene signatures at single-cell resolution

    • Identification of rare HORMAD1-expressing subpopulations within tumors

    • Trajectory analysis to track evolution of HORMAD1-expressing clones

  • Single-cell protein analysis:

    • Mass cytometry (CyTOF) to simultaneously quantify HORMAD1 and DNA repair proteins

    • Single-cell Western blotting for protein quantification in rare cells

    • Imaging mass cytometry for spatial context of HORMAD1-expressing cells

  • Single-cell DNA damage assessment:

    • CODEX multiplexed imaging to correlate HORMAD1 with multiple DNA damage markers

    • Single-cell comet assays to quantify DNA damage in HORMAD1-positive versus negative cells

    • γH2AX flow cytometry with HORMAD1 co-staining

  • Spatial transcriptomics:

    • Mapping HORMAD1 expression within the tumor microenvironment

    • Correlation with hypoxic regions that may induce replication stress

    • Integration with immune cell localization patterns

  • Lineage tracing:

    • Tracking the fate of HORMAD1-expressing cells during tumor evolution

    • Assessment of clonal advantage/disadvantage conferred by HORMAD1 expression

    • Correlation with treatment response at single-cell resolution

These approaches would help resolve how HORMAD1 can be protective in some cancer contexts (lung adenocarcinoma) while promoting genomic instability in others (triple-negative breast cancer) .

What are the most significant unresolved questions about HORMAD1's dual roles in meiosis and cancer?

Several critical questions remain unanswered regarding HORMAD1 biology:

  • Mechanistic divergence:

    • How does HORMAD1 promote interhomolog recombination in meiosis but affect sister chromatid recombination differently in cancer cells?

    • What post-translational modifications or binding partners redirect HORMAD1 function in cancer versus meiotic contexts?

  • Cancer type specificity:

    • Why does HORMAD1 promote genomic instability in breast cancer but protect against it in lung adenocarcinoma?

    • What molecular features determine whether HORMAD1 will be oncogenic or tumor-suppressive in a given context?

  • Therapeutic implications:

    • Can HORMAD1 expression be used as a biomarker for PARP inhibitor sensitivity beyond BRCA1/2 status?

    • Is HORMAD1 itself a viable therapeutic target, particularly in cancers where it promotes survival?

  • Evolutionary significance:

    • Why is HORMAD1 aberrantly expressed in multiple cancer types despite normally being restricted to meiotic cells?

    • Does HORMAD1 activation represent a specific adaptive response to genomic stress in cancer?

  • Regulatory mechanisms:

    • What triggers HORMAD1 expression in somatic cancer cells?

    • How is HORMAD1 recruited to different chromosomal contexts in cancer versus meiosis?

Resolving these questions will require integrated approaches combining structural biology, advanced imaging, genetic models, and clinical correlations. Particularly important will be determining the molecular switches that control HORMAD1's seemingly opposing functions in different cellular 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.