The METTL16 antibody is a specialized immunoglobulin designed to detect the methyltransferase-like protein 16 (METTL16), an N6-methyladenosine (m6A) RNA methyltransferase involved in RNA modification and gene regulation. It is widely used in molecular biology research to study METTL16’s role in cellular processes, including RNA splicing, mRNA stability, and cancer progression .
Reactivity: Primarily targets human, mouse, rat, and monkey METTL16 proteins .
Applications: Validated for Western blot (WB), immunohistochemistry (IHC), immunoprecipitation (IP), and enzyme-linked immunosorbent assay (ELISA) .
Sensitivity: Detects endogenous METTL16 in tissues and cell lysates .
Molecular Weight: Recognizes a ~64–78 kDa band, depending on post-translational modifications .
METTL16 contains three functional domains:
Methyltransferase Domain: Catalyzes m6A modifications on RNA targets (e.g., U6 snRNA, MAT2A mRNA) .
RNA-Binding Domains (RBD): Facilitates interactions with RNA substrates, including MALAT1 lncRNA .
Vertebrate Conserved Region (VCR): Mediates nuclear localization and RNA binding .
The antibody targets the METTL16 fusion protein Ag13889, ensuring specificity for the full-length protein . Cross-reactivity with METTL16 isoforms (64 kDa and 26 kDa) has been validated in immunoblotting .
Colorectal Cancer (CRC): METTL16 overexpression enhances tumor growth and immune evasion by stabilizing FBXO5 mRNA . Antibody-based studies revealed its role in PD-L1 regulation, a key immune checkpoint .
Breast Cancer (BC): METTL16 promotes malignancy by stabilizing oncogenic transcripts like FBXO5 . Its expression correlates with poor prognosis in BC patients .
Pancreatic Ductal Adenocarcinoma (PDA): High METTL16 levels predict favorable outcomes by enhancing tumor-infiltrating CD8+ T cells and naive B cells .
Hematopoietic Stem Cells (HSPCs): METTL16 is essential for cell cycle progression during early embryogenesis, as shown by single-cell RNA sequencing and antibody-based validation .
SAM Homeostasis: METTL16 acts as a metabolic sensor, regulating SAM levels via MAT2A mRNA splicing . Antibody-based knockdown experiments confirmed its role in maintaining SAM-dependent methylation .
Cross-Reactivity: Some antibodies (e.g., ab186012) show partial reactivity with METTL3/14 complexes, requiring careful validation .
Tissue Specificity: METTL16 is highly expressed in liver, kidney, and brain, necessitating optimized protocols for low-abundance tissues .
Epitope Masking: Denaturation methods (e.g., SDS-PAGE) may alter METTL16’s conformation, affecting antibody binding .
METTL16 antibodies are extensively utilized in several key applications within molecular biology and neuroscience research. Western blotting represents the most common application, with recommended dilutions typically ranging from 1:1000 to 1:10000 depending on the specific antibody and experimental conditions . Immunohistochemistry applications generally require dilutions between 1:500 to 1:2000, with optimal results achieved using TE buffer (pH 9.0) for antigen retrieval, though citrate buffer (pH 6.0) may serve as an alternative . For immunoprecipitation experiments, a 1:50 dilution has demonstrated effective results for pulling down endogenous METTL16 protein complexes . Additionally, these antibodies have proven valuable in RNA immunoprecipitation (RIP) assays to investigate direct interactions between METTL16 protein and target RNAs such as MAT2A mRNA .
Verifying antibody specificity is crucial for reliable results. A comprehensive validation approach should include:
Positive controls: Test the antibody on samples with known METTL16 expression. Validated positive controls include HeLa cells, NCI-H1299 cells, NIH/3T3 cells, and mouse/rat testis tissue .
Knockdown validation: Perform METTL16 knockdown experiments using shRNA or siRNA approaches. For example, lentiviral vectors (Lenti-sh-METTL16) or AAV-based systems (AAV-sh-METTL16) have demonstrated approximately 60% knockdown efficiency when measured by qRT-PCR and confirmed by western blotting .
Molecular weight confirmation: Verify detection at the expected molecular weight. METTL16 typically appears at 70-75 kDa on western blots, slightly higher than its calculated weight of 64 kDa (562 amino acids), likely due to post-translational modifications .
Cross-reactivity assessment: Most commercial METTL16 antibodies show reactivity with human, mouse, and rat samples; confirm specificity across your species of interest .
For robust validation of METTL16 antibodies, researchers should consider the following positive controls:
Cell lines:
HeLa cells (human cervical cancer)
NCI-H1299 cells (human non-small cell lung carcinoma)
NIH/3T3 cells (mouse fibroblasts)
MDA-MB-231 and MDA-MB-453 (breast cancer cell lines with confirmed METTL16 overexpression)
Tissue samples:
Mouse testis tissue (consistently shows high expression)
Rat testis tissue
Human colon tissue (for IHC applications)
Hippocampal tissue (particularly from MWM-trained mice, which show upregulated METTL16)
MCF10A cells (normal breast epithelial cells) can serve as a negative/low expression control in comparative studies with breast cancer cell lines .
Investigating direct METTL16-RNA interactions requires a multi-faceted approach:
RNA Immunoprecipitation (RIP): First, confirm the practicality of your anti-METTL16 antibody through western blotting. Then perform RIP by immunoprecipitating METTL16 protein complexes using anti-METTL16 antibody (with IgG as a negative control) and analyze captured RNA by qRT-PCR using primers specific to your target mRNA. For example, researchers successfully demonstrated METTL16 interaction with MAT2A mRNA using this approach, finding significant enrichment compared to IgG controls .
m6A RNA Immunoprecipitation (MeRIP): To confirm m6A modification of your target mRNA, perform MeRIP using anti-m6A antibodies followed by qRT-PCR for your target transcript. This approach revealed m6A modification of FBXO5 mRNA in breast cancer studies .
Methylated RNA stability assays: After confirming interaction, assess whether METTL16 affects mRNA stability through m6A modification. Treat cells with actinomycin D to inhibit transcription, then harvest RNA at different time points to determine mRNA half-life in METTL16-knockdown versus control conditions .
3'UTR reporter assays: For mapping precise interaction sites, clone the putative binding region (e.g., 3'UTR) into a reporter construct and assess expression in METTL16-knockdown versus control conditions .
To examine METTL16's function in learning and memory:
For challenging tissue samples:
Optimized tissue preparation:
For neural tissues (e.g., hippocampus), preserve tissue integrity through rapid extraction and flash-freezing
Consider perfusion fixation for IHC applications
For western blotting, include protease inhibitors and phosphatase inhibitors in lysis buffer
Enhanced antigen retrieval for IHC:
Signal amplification strategies:
For western blotting: Extend primary antibody incubation to overnight at 4°C
For IHC: Consider tyramide signal amplification systems
Use specialized detection systems (e.g., SuperSignal West Femto for WB)
Blocking optimization:
Test both BSA and milk-based blocking solutions
Consider species-specific protein blocks to reduce background
Include longer blocking steps (2+ hours) for high-background samples
When investigating METTL16's m6A methyltransferase function:
To differentiate direct from indirect effects:
Direct binding assessment:
m6A modification confirmation:
Mechanistic dissection:
Temporal analysis:
Time-course experiments following METTL16 manipulation
Early changes (0-4h) likely represent direct effects
Later changes (12-24h+) may include secondary/indirect effects
Troubleshooting recommendations:
Storage and handling optimization:
Protocol modifications:
Sample preparation refinement:
Ensure complete protein denaturation for WB
Verify protein extraction efficiency
For tissues with high protease activity, increase protease inhibitor concentration
Cross-validation:
When reconciling seemingly contradictory findings:
Context-specific regulation:
Target spectrum variation:
Pathway integration:
Examine downstream signaling pathways in each context
Consider interaction with other m6A regulatory proteins
Assess global m6A changes versus specific target effects
Technical considerations:
Confirm antibody specificity in each tissue type
Validate knockdown efficiency across different cell types
Consider potential compensatory mechanisms in different tissues
To explore METTL16's role in RNA stability:
Combined RIP and stability assays:
Site-specific mutation analysis:
Create constructs with wild-type or mutated METTL16 binding sites
Compare stability and expression levels between variants
Correlate with m6A levels at specific sites
m6A reader protein investigation:
Transcriptome-wide analysis:
Combine MeRIP-seq with RNA-seq in METTL16-knockdown versus control conditions
Correlate changes in m6A modification with abundance of transcripts
Identify common sequence motifs in stabilized/destabilized transcripts
Recent research has revealed that METTL16 is essential for embryonic development:
Developmental stage-specific analysis:
Stem cell differentiation studies:
Monitor METTL16 levels during stem cell differentiation using western blotting
Correlate with global m6A levels and differentiation markers
Investigate effects of METTL16 knockdown on lineage commitment
Transcriptome dysregulation assessment:
Compare transcriptome profiles between METTL16-deficient and wild-type embryonic cells
Identify key developmental genes regulated by METTL16-mediated m6A modification
Focus on pathways essential for early embryonic development
Methyltransferase activity during development:
Compare METTL16 activity between pluripotent and differentiated states
Identify developmental stage-specific RNA targets
Investigate interaction with developmental signaling pathways
To discover new METTL16 targets:
Integrated omics approach:
Perform RIP-seq using anti-METTL16 antibody to identify bound RNAs
Combine with MeRIP-seq to identify m6A-modified transcripts
Compare transcriptomes of METTL16-knockdown versus control cells
Look for RNAs that are both bound by METTL16 and show altered stability/abundance after knockdown
Structural motif analysis:
Analyze sequence and structural features of known targets (MAT2A, FBXO5, U6 snRNA)
Perform computational screening for similar motifs transcriptome-wide
Validate candidates experimentally using RIP and stability assays
Proximity labeling approaches:
Use METTL16 fusion proteins with proximity labeling enzymes
Identify RNAs in close proximity to METTL16 in living cells
Validate with traditional RIP using anti-METTL16 antibodies
Cross-linking immunoprecipitation (CLIP):
Perform CLIP-seq using anti-METTL16 antibodies
Map binding sites at nucleotide resolution
Correlate with m6A modification sites and RNA stability