Note: While we will prioritize shipping the format currently in stock, please specify your format preference in order notes. We will accommodate your request to the best of our ability.
Note: All proteins are shipped with standard blue ice packs. Dry ice shipping requires prior arrangement and incurs additional charges.
The tag type is determined during production. To ensure a specific tag, please inform us, and we will prioritize its development.
Recombinant Chicken Chromatin modification-related protein MEAF6 (MEAF6) is a component of the NuA4 histone acetyltransferase complex. This complex is involved in transcriptional activation of specific genes primarily through acetylation of nucleosomal histones H4 and H2A. This modification can alter nucleosome-DNA interactions and facilitate interactions with other transcriptionally-active proteins. MEAF6 is also a component of HBO1 complexes, specifically mediating acetylation of histone H3 at Lysine 14 (H3K14ac), exhibiting reduced activity towards histone H4. Furthermore, it is a component of the MOZ/MORF complex, possessing histone H3 acetyltransferase activity.
MEAF6 (MYST/Esa1-associated factor 6) is a chromatin modification-related protein that functions as part of histone acetyltransferase complexes. In chickens, MEAF6 is encoded on chromosome 23 and is also known by several synonyms including C1orf149, chromatin modification-related protein MEAF6, esa1-associated factor 6 homolog, and protein EAF6 homolog . The protein plays crucial roles in regulating gene expression through its involvement in chromatin remodeling complexes. For experimental work, researchers should note that recombinant forms typically maintain the core functional domains necessary for protein-protein interactions within these complexes.
Chicken MEAF6 shares significant homology with human MEAF6 (HGNC ID: HGNC:25674) , suggesting conserved functions across species. While zebrafish MEAF6 is available as a recombinant protein with >80% purity , the chicken variant may exhibit species-specific modifications or interaction patterns. Comparative analysis between chicken MEAF6 and human orthologs reveals important insights into evolutionary conservation of chromatin modification machinery. When designing cross-species experiments, researchers should account for potential functional differences by performing alignment analyses of protein domains prior to experimental design.
MEAF6 expression patterns in chicken tissues can be assessed through standard transcriptomic and proteomic approaches. The gene expression of chicken MEAF6 can be referenced through GEISHA (Gene Expression In Situ Hybridization Analysis) , which provides tissue-specific expression data during development. For experimental validation, researchers should employ tissue-specific RNA extraction followed by RT-qPCR with primers designed specifically for chicken MEAF6. Alternatively, immunohistochemistry using antibodies against conserved MEAF6 epitopes can reveal protein localization patterns across different tissues.
Based on experiences with other species' MEAF6 proteins, mammalian expression systems generally provide the most appropriate post-translational modifications for functional studies of chromatin-associated proteins. For chicken MEAF6, using a mammalian cell line similar to that used for zebrafish MEAF6 is recommended. HEK293 or CHO cells typically yield properly folded protein with relevant modifications. The expression construct should include a purification tag (His-tag is commonly used) and the chicken MEAF6 coding sequence optimized for the expression system. Transfection efficiency can be monitored through GFP co-expression, while protein expression levels should be validated by Western blot using tag-specific antibodies.
For optimal purification of recombinant chicken MEAF6, a multi-step approach is recommended. Based on related MEAF6 proteins, initial capture can be performed using affinity chromatography (Ni-NTA for His-tagged proteins) , followed by ion exchange chromatography to remove contaminants. Final polishing via size exclusion chromatography helps achieve >80% purity while maintaining native conformation. Throughout purification, protein activity should be monitored using functional assays such as histone binding or complex formation with known interaction partners. Storage in PBS buffer at -80°C with glycerol as a cryoprotectant helps maintain long-term stability .
Structural integrity verification for recombinant chicken MEAF6 requires a multi-faceted approach. Circular dichroism spectroscopy can confirm proper secondary structure elements, while limited proteolysis followed by mass spectrometry helps identify exposed regions versus protected domains. Thermal shift assays provide information on protein stability and proper folding. For functional verification, co-immunoprecipitation experiments with known interaction partners from chicken cell extracts should demonstrate native binding capabilities. Additionally, analytical size exclusion chromatography can reveal whether the protein exists in monomeric form or forms higher-order assemblies indicative of proper folding.
To study chicken MEAF6 interactions with chromatin complexes, chromatin immunoprecipitation (ChIP) followed by sequencing (ChIP-seq) provides genome-wide binding profiles. For this approach, researchers should use either a tagged recombinant MEAF6 protein introduced into chicken cell lines or develop chicken MEAF6-specific antibodies. Co-immunoprecipitation combined with mass spectrometry can identify novel protein-protein interactions within chromatin complexes. For in vitro interaction studies, recombinant MEAF6 can be immobilized on beads and used to pull down potential binding partners from nuclear extracts. Crosslinking experiments using formaldehyde or photo-activatable crosslinkers provide additional evidence for direct interactions in cellular contexts.
Assessment of chicken MEAF6 histone modification activities requires reconstitution of functional complexes, as MEAF6 typically functions as part of larger histone acetyltransferase complexes rather than acting independently. In vitro histone acetyltransferase (HAT) assays using recombinant chicken MEAF6 incorporated into its native complex should be performed with purified chicken histones or nucleosomes as substrates. Activity can be quantified by measuring acetylation levels using modification-specific antibodies in Western blots or by mass spectrometry. For cellular studies, researchers can introduce tagged MEAF6 into chicken cell lines and assess histone modification changes via ChIP-seq or immunofluorescence microscopy targeting specific histone marks.
To demonstrate chicken MEAF6 function in gene regulation, CRISPR-Cas9 mediated knockout or knockdown of MEAF6 in chicken cell lines (such as DT40 or primary cells) followed by RNA-seq analysis reveals global transcriptional impacts. For more targeted analysis, reporter gene assays using promoters of genes potentially regulated by MEAF6-containing complexes can be employed. Additionally, rescue experiments introducing recombinant chicken MEAF6 into MEAF6-depleted cells help confirm specificity of observed phenotypes. For dynamic studies, researchers should consider inducible expression systems to monitor temporal effects of MEAF6 activity on target gene expression through time-course experiments.
Alternative splicing of MEAF6 has been documented in other species, with significant functional consequences. For example, the MEAF6-1 splice variant in humans promotes cell proliferation and invasion in prostate cancer . To investigate potential alternative splicing in chicken MEAF6, researchers should perform RNA-seq on different chicken tissues and developmental stages, followed by RT-PCR validation using primers spanning potential splice junctions. Functional characterization of identified splice variants can be achieved by expressing each variant as a recombinant protein and comparing their interaction partners, chromatin binding profiles, and effects on target gene expression. Cell type-specific effects can be assessed by introducing each variant into various chicken cell lines and monitoring proliferation, differentiation, or other relevant phenotypes.
To investigate chicken MEAF6's role in development and differentiation, temporal expression analysis throughout embryonic development using in situ hybridization or tissue-specific RNA-seq provides initial insights. Functional studies can employ CRISPR-Cas9 to generate conditional MEAF6 knockout in specific tissues or at defined developmental timepoints. Ex vivo culture of chicken embryonic tissues with MEAF6 inhibition (via morpholinos or dominant-negative constructs) can reveal tissue-specific requirements. Comparison with known developmental phenotypes in other species helps identify conserved functions. Based on research in other contexts, researchers should pay particular attention to cell proliferation markers, as MEAF6 variants have been shown to regulate cell proliferation rates through pathways including ID1 and ID3 gene networks .
Post-translational modifications of chicken MEAF6 may significantly impact its function, similar to other chromatin-associated factors. To map these modifications, recombinant chicken MEAF6 can be expressed in chicken cell lines, immunoprecipitated, and analyzed by mass spectrometry. Potential phosphorylation, acetylation, methylation, or ubiquitination sites can be mutated to generate modified recombinant proteins for functional comparison. Phosphorylation-specific antibodies can be developed to monitor modification status under different cellular conditions or in response to signaling pathway activation. Changes in interaction partners or chromatin association following specific modifications provide important insights into regulatory mechanisms controlling MEAF6 function during different cellular processes.
When designing experiments with recombinant chicken MEAF6, several controls are essential. For protein interaction studies, researchers should include:
| Control Type | Purpose | Implementation |
|---|---|---|
| Negative Control | Exclude non-specific binding | Use purification tag only or irrelevant protein with same tag |
| Positive Control | Validate experimental system | Known interaction partner (e.g., components of HAT complexes) |
| Catalytically Inactive Mutant | Distinguish structural from enzymatic roles | Point mutations in key functional domains |
| Species Comparison Control | Assess evolutionary conservation | Human or mouse MEAF6 in parallel experiments |
| Domain Deletion Variants | Map functional regions | Systematic deletion of predicted domains |
Additionally, in cellular assays, appropriate vector-only controls and rescue experiments with wild-type protein should be performed to ensure specificity of observed phenotypes.
When encountering contradictory results in MEAF6 functional studies, researchers should systematically investigate potential sources of variation. First, consider whether different splice variants were studied, as variants like MEAF6-1 can have distinct functions from other isoforms . Second, evaluate cell type-specific effects, as MEAF6 may interact with different partners in various cellular contexts. Third, assess experimental conditions including protein concentration, buffer composition, and incubation times that might affect activity measurements. Establishing dose-response relationships rather than single-point measurements helps identify potential threshold effects. Finally, direct comparison experiments performed simultaneously under identical conditions provide the most reliable resolution of contradictory findings. Publication of negative results and comprehensive methods details facilitates community progress in resolving discrepancies.
Development of chicken MEAF6-specific antibodies requires careful epitope selection and validation. Researchers should:
Select unique epitopes by comparing chicken MEAF6 sequence with orthologs to identify both conserved regions (for cross-species reactivity) and unique regions (for species specificity)
Avoid regions prone to post-translational modifications that might block antibody recognition
Generate both monoclonal and polyclonal antibodies for complementary applications
Validate specificity using multiple approaches including Western blot against recombinant protein, immunoprecipitation followed by mass spectrometry, and reduced/absent signal in MEAF6 knockout cells
Test cross-reactivity against related family members and splice variants
Validate functionality in multiple applications (Western blot, immunoprecipitation, ChIP, immunofluorescence)
Proper antibody validation ensures reliable results and facilitates comparison across studies.
Analysis of ChIP-seq data for chicken MEAF6 binding patterns should follow a robust computational pipeline. After quality control and alignment to the chicken genome (bGalGal1.mat.broiler.GRCg7b or appropriate assembly) , peak calling using algorithms such as MACS2 identifies binding sites. Researchers should perform motif analysis to identify potential DNA binding motifs, though direct DNA binding might be limited as MEAF6 typically functions within protein complexes. Integration with histone modification data, especially acetylation marks, provides context for MEAF6 function. Comparison with gene expression data from the same cell types helps associate binding events with transcriptional outcomes. Differential binding analysis between experimental conditions (e.g., development stages, cell types) reveals context-specific functions. Gene ontology and pathway enrichment analyses of MEAF6-associated genes provide functional insights into biological processes regulated by this chromatin modifier.
When analyzing functional effects of chicken MEAF6, appropriate statistical approaches depend on experimental design. For comparison of gene expression changes, differential expression analysis tools such as DESeq2 or edgeR with appropriate multiple testing correction (FDR < 0.05) should be applied. For phenotypic assays (e.g., proliferation, differentiation), ANOVA followed by post-hoc tests is appropriate when comparing multiple conditions, while t-tests (paired or unpaired depending on experimental design) can be used for binary comparisons. Power analysis should be performed prior to experimentation to determine appropriate sample sizes. For time-course experiments, repeated measures ANOVA or mixed-effects models account for temporal correlation. Integration of multiple data types may require multivariate approaches such as principal component analysis or partial least squares regression to identify patterns across datasets.
Distinguishing direct from indirect effects of chicken MEAF6 in omics datasets requires integration of multiple experimental approaches. Direct targets can be identified by combining ChIP-seq binding data with rapid transcriptional responses following acute MEAF6 modulation (e.g., using degradation tag systems or inducible expression). Time-course experiments following MEAF6 activation or inhibition help establish temporal order of effects, with early changes more likely representing direct targets. Motif analysis at binding sites can identify potential co-factors mediating MEAF6 genomic recruitment. Analysis of protein complexes containing MEAF6 through proximity labeling approaches (BioID, APEX) helps identify direct protein interactions. Network analysis identifying first-neighbor versus higher-order connections in regulatory networks can further classify direct versus indirect effects. Validation experiments using reporter assays for selected targets provide confirmatory evidence for direct regulation.
Future research on chicken MEAF6 should focus on several promising directions. First, comprehensive characterization of potential splice variants similar to the functionally distinct MEAF6-1 variant identified in human cells may reveal tissue-specific functions in chickens. Second, investigation of MEAF6's role in development using chicken embryo models could provide insights into evolutionarily conserved chromatin regulation mechanisms. Third, exploring MEAF6's function in immune cell development and function in chickens may uncover species-specific adaptations. Fourth, structural biology approaches to determine MEAF6's three-dimensional structure and its conformation within different complexes would advance understanding of its molecular function. Finally, development of small molecule inhibitors or modulators of MEAF6 function would provide valuable tools for dissecting its roles in various cellular processes and potentially reveal therapeutic applications for conditions involving dysregulated chromatin modification.