ZMYM3 antibodies vary in clonality, host species, and applications. Key commercial antibodies include polyclonal and monoclonal variants with distinct reactivity profiles.
Polyclonal vs. Monoclonal: Polyclonal antibodies (e.g., A08961, 25742-1-AP) detect multiple epitopes, offering broader reactivity, while monoclonal antibodies (e.g., C15200016) provide higher specificity.
Reactivity: Most antibodies target human ZMYM3, with cross-reactivity to mouse in A08961 and 25742-1-AP .
Observed Molecular Weight: Varies between 68 kDa (A08961) and 190–200 kDa (25742-1-AP), likely due to isoforms or post-translational modifications .
ZMYM3 antibodies enable diverse experimental approaches, including chromatin studies, protein-protein interactions, and oncology research.
ZMYM3 binds chromatin and DNA, facilitating BRCA1-A complex recruitment to DNA double-strand breaks (DSBs) . Antibodies have validated these interactions:
ChIP-seq: Diagenode’s C15200016 detects ZMYM3 enrichment at DSBs, linking it to homologous recombination (HR) repair .
Immunofluorescence: A08961 and ab106626 localize ZMYM3 to nuclear foci following DNA damage, confirming its role in chromatin remodeling .
ZMYM3 promotes cell proliferation in small cell lung carcinoma (SCLC):
Knockdown Experiments: ZMYM3 depletion reduces SCLC cell growth (e.g., SBC-3 line), decreases Ki-67 labeling, and delays tumor formation in xenograft models .
Immunoblotting: NovoPro’s 161376 and Proteintech’s 25742-1-AP detect ZMYM3 expression in lung cancer cell lines, correlating with cyclin D1 levels .
While ZMYM3 antibodies are research tools, their findings highlight therapeutic potential:
Isoform Variability: Antibodies may detect distinct ZMYM3 isoforms, necessitating validation for specific studies .
Species-Specific Reactivity: Most antibodies target human ZMYM3; cross-reactivity with non-human models requires testing .
Functional Studies: Further work is needed to dissect ZMYM3’s dual role in DNA repair and oncogenesis, leveraging CRISPR-based models.
ZMYM3 (Zinc finger MYM-type protein 3, also known as ZNF261, DXS6673E, or KIAA0385) is a chromatin-binding protein with significant roles in DNA damage response pathways. It functions as a critical factor in modulating BRCA1 functions within chromatin to maintain genome integrity . ZMYM3 is a 1370-amino-acid protein containing 10 tandem zinc finger domains along with a domain of unknown function (DUF3504) . The protein plays essential roles in:
DNA repair through homologous recombination (HR) mechanisms
Spermatogenesis in males, with knockout studies showing arrest at the metaphase of the first meiotic division
Research interest in ZMYM3 has increased due to its implications in genome stability pathways and potential connections to disease mechanisms when dysregulated.
Based on validated research applications, ZMYM3 antibodies are most suitable for:
Western blotting (WB): Detecting ZMYM3 protein in nuclear extracts, with expected band sizes around 152 kDa for the primary isoform , though multiple isoforms have been observed at approximately 200 kDa and 95 kDa
Immunohistochemistry on paraffin-embedded sections (IHC-P): Visualizing ZMYM3 distribution in tissue sections
Immunoprecipitation (IP): Particularly useful for studying protein-protein interactions, such as the association between ZMYM3 and LSD1
Immunofluorescence: For examining subcellular localization patterns, which vary between isoforms (nuclear localization for the larger isoform, both nuclear and cytoplasmic distribution for the smaller isoform)
When selecting an antibody for these applications, researchers should verify specificity for the target region of interest, as alternative splicing may generate multiple ZMYM3 isoforms with differing functional domains.
Proper validation of ZMYM3 antibodies requires a multi-faceted approach:
Knockout/knockdown controls: Compare antibody signals between wild-type cells and ZMYM3 knockout or knockdown models. Complete loss of signal in knockout samples provides strong evidence of specificity
Western blot analysis: Verify band patterns match predicted molecular weights, acknowledging that post-translational modifications often result in migration patterns different from theoretical weights. For ZMYM3, expect bands at approximately 152 kDa theoretical weight, though observed bands of ~200 kDa and 95 kDa have been reported due to post-translational modifications
Peptide competition assay: Pre-incubate the antibody with the immunizing peptide (synthetic peptide within Human ZMYM3 aa 250-300 for some commercial antibodies) before application in your experimental system. Signal elimination confirms epitope-specific binding
Subcellular fractionation correlation: Confirm that detected signals align with expected subcellular distribution patterns. For ZMYM3, the larger isoform (~200 kDa) should be predominantly nuclear, while the smaller isoform (~95 kDa) should be detected in both nuclear and cytoplasmic fractions
Cross-reference multiple antibodies: Where possible, compare results using different antibodies targeting distinct ZMYM3 epitopes to confirm consistent patterns
Detecting ZMYM3 in chromatin fractions requires careful optimization due to its tight association with both histone and DNA components of nucleosomes. The following protocol has been validated in research settings:
Chromatin Fractionation Protocol for ZMYM3 Detection:
Cell preparation: Harvest approximately 1-5×10⁶ cells and wash twice with ice-cold PBS
Cytoplasmic extraction:
Resuspend cell pellet in 200 μl cytoplasmic extraction buffer (10 mM HEPES pH 7.9, 10 mM KCl, 1.5 mM MgCl₂, 0.34 M sucrose, 10% glycerol, 1 mM DTT, protease inhibitors)
Add Triton X-100 to 0.1% final concentration
Incubate on ice for 8 minutes
Centrifuge at 1,300 × g for 5 minutes at 4°C
Collect supernatant as cytoplasmic fraction
Nuclear extraction:
Wash nuclear pellet once with cytoplasmic extraction buffer
Resuspend in 200 μl nuclear extraction buffer (3 mM EDTA, 0.2 mM EGTA, 1 mM DTT, protease inhibitors)
Incubate on ice for 30 minutes with occasional vortexing
Centrifuge at 1,700 × g for 5 minutes at 4°C
Collect supernatant as nuclear soluble fraction
Chromatin extraction:
Resuspend pellet in 200 μl chromatin extraction buffer (50 mM Tris-HCl pH 7.5, 150 mM NaCl, 1% NP-40, 0.5% sodium deoxycholate, 0.1% SDS, 1 mM DTT, benzonase 250 U/ml, protease inhibitors)
Incubate at room temperature for 15 minutes with rotation
Centrifuge at 16,000 × g for 10 minutes
Western blot detection:
Research has demonstrated that endogenous ZMYM3 is exclusively present in the chromatin fraction, making this extraction protocol particularly important for successful detection .
ChIP-seq for ZMYM3 requires special considerations due to its bivalent interactions with both histone and DNA components of nucleosomes. The following optimized protocol incorporates techniques from published ZMYM3 ChIP-seq studies:
Cell preparation:
Crosslinking and chromatin preparation:
Double crosslinking approach is recommended: first with 2 mM disuccinimidyl glutarate (DSG) for 45 minutes, followed by 1% formaldehyde for 10 minutes at room temperature
Quench with 0.125 M glycine for 5 minutes
Wash cells twice with ice-cold PBS
Lyse cells and isolate nuclei using SDS lysis buffer
Sonicate chromatin to 200-500 bp fragments
Immunoprecipitation optimization:
Pre-clear chromatin with protein A/G beads
Use 5-10 μg of validated ZMYM3 antibody per ChIP reaction
Include appropriate controls: IgG negative control and H3K4me3 positive control
Incubate overnight at 4°C with rotation
Epitope tagging approach:
Bioinformatic analysis:
This optimized protocol accounts for ZMYM3's unique chromatin binding properties through double crosslinking and considers the alternative CETCh-seq approach for challenging epitopes.
Multiple ZMYM3 bands in Western blots reflect biologically significant isoforms rather than non-specific binding. Proper interpretation requires understanding the following key points:
Expected band patterns:
Isoform-specific subcellular distribution:
Alternative splicing considerations:
Troubleshooting inconsistent band patterns:
Issue | Potential Cause | Solution |
---|---|---|
Missing larger (200 kDa) band | Insufficient nuclear extraction | Ensure complete nuclear lysis; use appropriate extraction buffers with nuclease treatment |
Missing smaller (95 kDa) band | Exposure time too short | Increase exposure time up to 15 minutes |
Additional unexpected bands | Cross-reactivity with related proteins | Perform peptide competition assay to identify specific bands |
Smeared bands | Protein degradation | Add fresh protease inhibitors; maintain cold temperatures throughout extraction |
Functional interpretation:
When reporting ZMYM3 Western blot results, clearly specify which isoform(s) you are detecting and include molecular weight markers to facilitate interpretation by other researchers.
Selecting appropriate cell models is critical for investigating ZMYM3's role in DNA damage response pathways. Based on published research, the following models offer distinct advantages:
HeLa cells:
U2OS cells:
HEK293T cells:
Spermatogonial stem cells (SSCs):
Physiologically relevant for studying ZMYM3 in reproductive contexts
Express both ZMYM3 isoforms with distinct subcellular localization patterns
Allow investigation of ZMYM3 interaction with LSD1 and regulation by growth factors
More challenging to culture but provide insights into tissue-specific functions
ZMYM3 knockout models:
For comprehensive characterization, a multi-model approach is recommended, starting with established cell lines for mechanistic studies and progressing to more specialized models for context-specific functions.
Analyzing the interaction between ZMYM3 and the BRCA1-A complex requires specialized approaches due to the dynamic nature of these interactions, particularly in the context of DNA damage. The following methodological approach is recommended:
Co-immunoprecipitation (Co-IP) studies:
Prepare nuclear extracts from cells with and without DNA damage induction
Use anti-ZMYM3 antibody for immunoprecipitation, followed by Western blotting for BRCA1-A components (ABRA1, RAP80)
Alternatively, perform reverse Co-IP using antibodies against BRCA1-A components
Include appropriate controls: IgG negative control and input samples
Proximity ligation assay (PLA):
This technique allows visualization of protein-protein interactions in situ
Fix cells with 4% paraformaldehyde after appropriate treatments
Perform PLA using antibody pairs targeting ZMYM3 and individual BRCA1-A components
Quantify PLA signals in different cellular compartments and in response to DNA damage
Sequential ChIP (ChIP-reChIP):
Perform first round of ChIP using ZMYM3 antibody
Elute complexes and perform second round using antibodies against BRCA1-A components
This approach identifies genomic regions where both proteins co-localize
FRET-based interaction analysis:
Generate fluorescently tagged ZMYM3 and BRCA1-A components
Measure FRET efficiency before and after DNA damage induction
This approach provides spatiotemporal information about interaction dynamics
Deletion mutant analysis:
Generate deletion constructs of ZMYM3 based on domain organization
Focus on the DNA-binding domain (amino acids 300-330) and chromatin-interacting regions
Perform Co-IP experiments with each mutant to map interaction domains
Complement with functional assays to correlate binding with biological activities
This multi-method approach provides comprehensive characterization of the ZMYM3-BRCA1-A interaction, which is critical for understanding ZMYM3's role in facilitating BRCA1 localization to damaged chromatin and promoting DNA repair by homologous recombination .
Rigorous experimental controls are critical when investigating ZMYM3 recruitment to DNA damage sites to ensure valid and reproducible results. The following controls should be incorporated:
Antibody specificity controls:
DNA damage induction controls:
Untreated control cells to establish baseline ZMYM3 distribution
Positive control proteins with established damage recruitment patterns (e.g., γH2AX, 53BP1)
Time course analysis to capture transient recruitment dynamics
Different DNA damage agents to distinguish damage-type specificity:
Domain-specific recruitment controls:
Cell cycle controls:
Synchronize cells in different cell cycle phases
Co-stain with cell cycle markers (e.g., PCNA for S-phase)
ZMYM3 recruitment may vary depending on cell cycle stage due to the availability of homologous recombination repair pathways
Quantification controls:
Establish clear criteria for positive recruitment (e.g., >1.5-fold enrichment at damage sites)
Perform quantification on sufficient numbers of cells (minimum 50-100 cells per condition)
Conduct statistical analysis to determine significance of recruitment
Use objective image analysis software to avoid confirmation bias
By implementing these controls, researchers can confidently assess ZMYM3 recruitment to DNA damage sites and make valid comparisons between experimental conditions, thereby advancing understanding of ZMYM3's role in the DNA damage response.
ZMYM3 exhibits variable expression patterns across different tissues, which can lead to seemingly contradictory results. To properly interpret and reconcile these differences, researchers should consider the following methodological approach:
Tissue-specific expression profiling:
Isoform-specific analysis:
Developmental timing considerations:
Regulatory influences:
Reconciliation strategy for contradictory results:
Observation | Possible Explanation | Reconciliation Approach |
---|---|---|
Different band patterns on Western blots | Tissue-specific isoform expression | Use multiple antibodies targeting different epitopes; include tissue-specific positive controls |
Varying subcellular localization | Cell type-specific regulation of nuclear import/export | Perform careful subcellular fractionation; use consistent cell fixation methods |
Discrepancies between mRNA and protein levels | Post-transcriptional regulation | Measure both parameters in the same samples; investigate miRNA regulation |
Inconsistent immunostaining patterns | Epitope masking in certain contexts | Test multiple antibodies and fixation protocols; include appropriate controls |
By systematically addressing these factors, researchers can develop a more comprehensive understanding of ZMYM3 biology that accounts for tissue-specific variations while establishing core functional principles.
Interpreting ZMYM3 knockout phenotypes requires careful consideration of several factors that could lead to misinterpretation. Researchers should be aware of these potential pitfalls and implement appropriate controls:
Sex-specific effects:
Compensatory mechanisms:
Acute versus chronic loss of ZMYM3 may yield different phenotypes
Consider using inducible knockout systems to distinguish between direct effects and adaptive responses
Examine expression changes in related family members (other ZMYM proteins) that might compensate for ZMYM3 loss
Cell type-specific functions:
Methodological considerations:
Complete versus partial knockouts may yield different phenotypes
Verify knockout efficiency at both mRNA and protein levels using isoform-specific detection methods
For CRISPR-Cas9 generated knockouts, check for potential off-target effects
Interpretation framework for complex phenotypes:
Molecular mechanism validation:
Correlate phenotypes with specific molecular functions of ZMYM3
For DNA repair defects, assess specifically homologous recombination efficiency
For meiotic phenotypes, examine spindle assembly and chromosome segregation
Rescue experiments with wild-type and mutant ZMYM3 are essential for causality determination
By addressing these considerations, researchers can develop more accurate interpretations of ZMYM3 knockout phenotypes across different experimental systems.
Resolving contradictory findings regarding ZMYM3's role in epigenetic regulation requires carefully designed experiments that address specific aspects of this complex function. The following methodological approach can help reconcile apparently contradictory results:
By implementing this comprehensive approach, researchers can develop a more nuanced understanding of ZMYM3's epigenetic functions that accounts for context-specific effects and resolves apparent contradictions in the literature.
Several cutting-edge technologies are particularly well-suited for investigating ZMYM3's complex roles in chromatin dynamics and DNA damage response. Researchers should consider these approaches to advance beyond current knowledge limitations:
Proximity labeling technologies:
Advanced microscopy techniques:
Super-resolution microscopy (STORM, PALM, SIM) to visualize ZMYM3 localization at the nanoscale
Live-cell imaging with lattice light-sheet microscopy to track ZMYM3 dynamics during DNA damage responses
Single-molecule tracking to analyze ZMYM3 mobility on chromatin before and after DNA damage
Genomic technologies:
CUT&Tag for improved mapping of ZMYM3 genomic binding sites with lower background
HiChIP to connect ZMYM3 binding with 3D chromatin structure
Single-cell technologies to address cellular heterogeneity:
scRNA-seq to identify cell state-specific transcriptional effects
scATAC-seq to link ZMYM3 to chromatin accessibility changes at single-cell resolution
CRISPR-based screening approaches:
CRISPR activation/inhibition screens targeting ZMYM3-regulated genes
CRISPR tiling screens across the ZMYM3 locus to identify functional regulatory elements
Base editing to introduce specific point mutations for structure-function analysis
Structural biology advances:
Cryo-EM studies of ZMYM3 in complex with nucleosomes and DNA repair machinery
Hydrogen-deuterium exchange mass spectrometry to map protein-protein interaction surfaces
AlphaFold2-based structural predictions to guide targeted mutagenesis of key domains
Multi-omics integration platforms:
Technology | Application to ZMYM3 Research | Expected Advancement |
---|---|---|
Spatial transcriptomics | Map ZMYM3-dependent gene expression in tissue context | Connect molecular mechanisms to physiological roles |
Proteomics with PTM analysis | Identify ZMYM3 modifications and their regulatory role | Understand activation/inactivation mechanisms |
Metabolomics integration | Link ZMYM3 function to cellular metabolic states | Discover unexpected connections to cellular physiology |
4D Nucleome approaches | Connect ZMYM3 to chromatin architecture changes over time | Understand higher-order regulatory functions |
These emerging technologies promise to resolve current knowledge gaps regarding ZMYM3's precise molecular mechanisms, tissue-specific functions, and potential roles in disease processes, guiding more targeted therapeutic interventions for conditions involving dysregulated ZMYM3.
Designing experiments to distinguish and characterize the functions of different ZMYM3 isoforms requires a systematic approach that combines molecular, cellular, and genomic techniques. The following experimental design strategy addresses this complex question:
Comprehensive isoform identification and characterization:
Perform long-read RNA sequencing (Oxford Nanopore or PacBio) to identify all ZMYM3 transcript isoforms
Compare isoform expression across tissues, with focus on brain and testis where ZMYM3 is highly expressed
Develop isoform-specific RT-PCR assays for quantitative analysis
Create a reference map of protein domains present in each isoform
Isoform-specific antibody development and validation:
Isoform-specific knockout/knockin strategies:
Design CRISPR-Cas9 editing strategies that target specific exons unique to individual isoforms
Create cell lines expressing only one isoform at a time
Generate knockin models with epitope-tagged versions of each isoform for tracking
Comparative functional studies:
Domain swap experiments:
Create chimeric constructs that exchange domains between isoforms
Test these constructs in rescue experiments with ZMYM3 knockout cells
This approach can identify which domains confer isoform-specific functions
Regulation of isoform expression:
Physiological significance assessment:
Analyze isoform expression in response to cellular stresses (DNA damage, oxidative stress)
Examine isoform ratios in relevant disease models
Correlate isoform expression with functional outcomes in tissue-specific contexts
This comprehensive experimental approach will provide crucial insights into the distinct biological roles of ZMYM3 isoforms, potentially explaining the diverse functions attributed to this protein across different cellular contexts.