MORF4L2 is a component of the NuA4 histone acetyltransferase complex involved in transcriptional activation of select genes principally by acetylation of nucleosomal histone H4 and H2A . This modification alters nucleosome-DNA interactions and promotes interaction of modified histones with transcription-regulating proteins. Research indicates MORF4L2 is critical for the activation of transcriptional programs associated with:
Oncogene and proto-oncogene mediated growth induction
Tumor suppressor mediated growth arrest
Replicative senescence
Apoptosis mechanisms
DNA repair processes
MORF4L2 antibodies enable researchers to detect this protein in various experimental contexts through multiple techniques including Western blotting, immunofluorescence, immunohistochemistry, and ELISA. These methods allow visualization of MORF4L2's localization (primarily nuclear), measurement of expression levels, and investigation of protein-protein interactions within the NuA4 complex .
MORF4L2 has a calculated molecular weight of approximately 32 kDa, but the observed molecular weight in Western blot applications typically falls between 30-32 kDa . This slight discrepancy between calculated and observed weights may be attributed to post-translational modifications or protein folding characteristics. When conducting Western blot analysis with MORF4L2 antibodies, researchers should expect to observe bands in this range when using appropriate reducing conditions and separation techniques .
The antibody dilution recommendations for Western blot applications generally range from 1:500 to 1:1000, though this may vary between specific antibody products . Positive Western blot detection has been confirmed in various cell lines including HeLa, A431, A549, and MCF-7 cells, as well as in human and mouse liver tissues .
Cross-reactivity of MORF4L2 antibodies varies based on the specific product and immunogen used. Most commercially available antibodies show high reactivity with human MORF4L2 samples . Many antibodies also demonstrate cross-reactivity with mouse and rat MORF4L2, making them suitable for comparative studies across these common model organisms .
Sequence homology analysis through BLAST reveals the following cross-reactivity potential by species:
Human: 100% identity
Mouse, Rat, Dog, Horse: approximately 92% identity
Bovine: 84% identity
Other species (Pig, Rabbit, Guinea Pig): variable identity ranging from 84-92%
When selecting a MORF4L2 antibody for cross-species applications, researchers should verify the specific cross-reactivity profile of their chosen antibody and consider sequence homology in the immunogen region.
Recent research has revealed significant associations between MORF4L2 expression and cancer progression. Studies indicate that high expression of MORF4L2 is associated with worse clinical features and increased macrophage infiltration in breast cancer tissues . Specifically, MORF4L2 has been found to induce immunosuppressive microenvironments and contribute to immunotherapy resistance through the GRHL2/MORF4L2/H4K12Ac/CSF1 axis in triple-negative breast cancer .
Correlation analyses have demonstrated that MORF4L2 expression exhibits:
Strong negative association with CD8+ T cells
Positive correlation with M2 macrophages and resting mast cells
Consistent negative correlation with CD8+ T cells alongside other progression-related DNA damage repair genes (GNPNAT1 and VAV3)
Immunohistochemistry sections have shown significant co-localization of positively stained regions of MORF4L2 with CD68, a marker of macrophages, while few CD8+ T cells were detected within these regions . Functional studies have revealed that knockdown of MORF4L2 resulted in impaired proliferation of MCF-7 breast cancer cells, suggesting its direct role in promoting cancer cell growth .
Research has uncovered an important inverse relationship between MORF4L2 and miR-3156-5p expression in the context of multiple endocrine neoplasia type 1 (MEN1). Studies have shown that miR-3156-5p is significantly downregulated in MEN1 patients compared to matched unaffected relatives .
In vitro functional studies in BON-1 pancreatic neuroendocrine tumor cells demonstrated that:
Transfection with miR-3156-5p mimic significantly reduced MORF4L2 protein expression by 46% (p<0.005)
Treatment with miR-3156-5p inhibitor significantly increased MORF4L2 expression 1.5-fold (p<0.05)
These findings suggest that miR-3156-5p directly regulates MORF4L2 expression, and this relationship may serve as a potential serum biomarker for neuroendocrine tumor occurrence in MEN1 patients. This inverse relationship contributes to the reliability of non-invasive biomarker approaches for monitoring tumor development and progression .
Validating antibody specificity is crucial for ensuring reliable research results. For MORF4L2 antibodies, a multi-faceted validation approach is recommended:
Positive and negative control samples: Use cell lines with known MORF4L2 expression levels. HeLa, MCF-7, and A549 cells are documented to express MORF4L2 and serve as positive controls .
Recombinant protein validation: Many suppliers provide recombinant MORF4L2 protein fragments (PrEST Antigens) that can be used as positive controls in Western blot applications .
siRNA knockdown validation: Transfect cells with MORF4L2-specific siRNAs to create a negative control. Studies have shown successful gene silencing of MORF4L2 in MCF-7 cells, resulting in reduced expression that can be verified by antibody-based detection methods .
Cross-validation with multiple antibodies: Use antibodies targeting different epitopes of MORF4L2 to confirm that the observed signal is specific to the target protein rather than cross-reactivity with other proteins.
Peptide competition assay: Pre-incubate the antibody with the immunizing peptide prior to application in the detection method. If the signal disappears or is significantly reduced, this supports antibody specificity .
Optimizing experimental conditions for MORF4L2 detection varies by application:
Sample preparation: Complete cell lysis in RIPA buffer with protease inhibitors
Secondary antibody: Anti-rabbit IgG (for rabbit-hosted primary antibodies)
Fixation: 4% paraformaldehyde for 15 minutes at room temperature
Permeabilization: 0.1-0.5% Triton X-100 for 10 minutes
Blocking: 1-5% BSA or normal serum
Nuclear counterstain: DAPI recommended due to nuclear localization of MORF4L2
Fixation: Formalin-fixed, paraffin-embedded tissues
Antigen retrieval: Heat-induced epitope retrieval in citrate buffer (pH 6.0)
Blocking: Endogenous peroxidase blocking with 3% H₂O₂ followed by protein blocking
Detection systems: HRP-polymer conjugated secondary antibodies with DAB substrate
Counterstain: Hematoxylin for nuclear contrast
Robust experimental design for MORF4L2 antibody applications should include several types of controls:
Cell lines with confirmed MORF4L2 expression (HeLa, MCF-7, A549, A431)
Recombinant MORF4L2 protein fragments (for Western blot applications)
Primary antibody omission control
Isotype control (rabbit IgG for rabbit-hosted primary antibodies)
Non-expressing tissues or cell lines (tissue-specific, must be validated)
Loading control for Western blot (e.g., tubulin, GAPDH, or β-actin)
Internal control for IHC (tissue with known expression pattern)
Background fluorescence control for IF/ICC
Multiple antibodies targeting different MORF4L2 epitopes
Correlation of protein detection with mRNA expression data
Peptide competition assay with immunizing peptide
When investigating MORF4L2's interactions with other proteins, particularly components of the NuA4 complex or in cancer research contexts, the following methodological approaches are recommended:
Use antibodies raised in different host species (e.g., rabbit anti-MORF4L2 and mouse anti-CD68)
Apply primary antibodies sequentially or simultaneously (protocol-dependent)
Use spectrally distinct fluorophore-conjugated secondary antibodies
Include single-stained controls to assess bleed-through
Quantify co-localization using correlation coefficients (e.g., Pearson's, Manders')
More sensitive for detecting protein-protein interactions
Provides higher spatial resolution than conventional co-localization
Generates punctate signals only when proteins are within 40nm proximity
Useful for confirming direct interactions between MORF4L2 and other NuA4 complex components
Use MORF4L2 antibodies for pull-down experiments
Western blot for interacting proteins or mass spectrometry for unbiased detection
Include IgG control immunoprecipitation
Consider crosslinking for transient interactions
Research has shown successful co-localization studies between MORF4L2 and immune cell markers such as CD68 (macrophage marker), demonstrating the utility of these techniques for investigating MORF4L2's role in modulating the tumor microenvironment .
Solution: Optimize antibody dilution (start with 1:500-1:1000)
Increase blocking time/concentration (5% milk or BSA for 2 hours)
Try different blocking agents (switch between milk and BSA)
Increase washing frequency and duration
Use gradient gels for better separation around 30-32 kDa region
Solution: Optimize antigen retrieval methods (try both citrate and EDTA buffers)
Extend primary antibody incubation (overnight at 4°C)
Use signal amplification systems (biotin-streptavidin or tyramide)
Test different fixation protocols for better epitope preservation
Solution: Increase blocking time/concentration
Add 0.1-0.3% Triton X-100 to antibody diluent
Pre-absorb primary antibody with tissue powder
Use more stringent washing (0.1% Tween-20 in PBS)
Try fluorophore-conjugated Fab fragments instead of whole IgG secondaries
Solution: Optimize fixation time for each tissue type
Adjust antigen retrieval time based on tissue density
Use tissue-specific blocking agents to reduce non-specific binding
Consider using automated staining platforms for consistency
Validate antibody performance in each tissue type independently
Discrepancies between MORF4L2 protein and mRNA expression are not uncommon and may reflect important biological processes. Consider these interpretive approaches:
Post-transcriptional regulation: Research has demonstrated that miR-3156-5p directly regulates MORF4L2 protein expression without necessarily affecting mRNA levels . When interpreting discrepancies, consider:
Analyzing miRNA expression profiles, particularly miR-3156-5p
Examining mRNA stability through actinomycin D chase experiments
Investigating RNA-binding proteins that might regulate MORF4L2 mRNA
Protein stability differences: MORF4L2 protein stability may vary across:
Different cell types or tissue contexts
Normal versus pathological states
Response to treatment conditions
Investigate using proteasome inhibitors (MG132) to assess degradation rates
Technical considerations:
Biological relevance:
Recent studies have revealed significant correlations between MORF4L2 expression and immune cell infiltration patterns in cancer tissues . To effectively integrate these datasets:
Sequential or multiplex staining approaches:
Computational analysis integration:
Functional validation experiments:
Single-cell analysis approaches:
Perform single-cell RNA-seq to correlate MORF4L2 expression with immune cell states
Use CyTOF or single-cell proteomics to examine protein-level correlations
Integrate spatial transcriptomics to map MORF4L2 expression in the tumor microenvironment
Recent findings demonstrate that MORF4L2 may induce immunosuppressive microenvironments through specific molecular pathways, particularly in triple-negative breast cancer . Understanding these relationships can help develop more effective immunotherapy strategies and identify patients who might benefit from targeted approaches.
MORF4L2 shows promise as a biomarker in several disease contexts:
High MORF4L2 expression correlates with worse clinical features in breast cancer
Part of a DNA damage-repair-related signature that reflects microenvironmental features and therapeutic response
May predict response to immunotherapy through its role in modulating immune cell infiltration
Knockdown of MORF4L2 impairs proliferation of cancer cells, suggesting therapeutic targeting potential
The inverse relationship between miR-3156-5p and MORF4L2 represents a potential serum biomarker for NET occurrence in MEN1 patients
This relationship could facilitate the detection of NET development through non-invasive blood testing
The combination of these markers may improve sensitivity and specificity compared to single markers
Validate expression in larger patient cohorts using tissue microarrays
Develop serum-based detection methods for circulating MORF4L2
Integrate MORF4L2 with other markers for improved diagnostic accuracy
Correlate expression with treatment response and clinical outcomes
Establish standardized detection protocols for clinical implementation
As a component of the NuA4 histone acetyltransferase complex, MORF4L2's role in histone modification is being investigated using advanced techniques:
ChIP-seq to map genome-wide binding sites of MORF4L2
CUT&RUN for higher resolution mapping with lower background
HiChIP to connect MORF4L2 binding with 3D chromatin organization
Re-ChIP to identify genomic regions bound by multiple NuA4 components
ChIP-seq for H4K12Ac and other relevant histone marks in MORF4L2-modulated systems
Mass spectrometry to characterize histone modification patterns
Antibody-based detection of specific modifications (Western blot, IF)
ATAC-seq to assess chromatin accessibility changes related to MORF4L2 function
CRISPR-Cas9 screening to identify genetic dependencies on MORF4L2
CRISPRi/CRISPRa for precise modulation of MORF4L2 expression
RNA-seq to characterize transcriptional programs regulated by MORF4L2
proteomics to identify MORF4L2 interaction partners and complex components
Cryo-EM to resolve structure of MORF4L2 within the NuA4 complex
Hydrogen-deuterium exchange mass spectrometry for protein dynamics
Cross-linking mass spectrometry to map protein-protein interactions
Recent research has identified the GRHL2/MORF4L2/H4K12Ac/CSF1 axis as a mechanism through which MORF4L2 induces immunosuppressive microenvironments in triple-negative breast cancer , highlighting the importance of understanding MORF4L2's role in histone modification for therapeutic development.