MYL9 is a myosin regulatory light chain with a canonical length of 172 amino acid residues and a molecular weight of approximately 19.8 kDa. It functions as a regulatory subunit that plays a crucial role in both smooth muscle and nonmuscle cell contractile activity through its phosphorylation state. MYL9 is primarily localized in the cytoplasm and exists in two alternatively spliced isoforms. It's a critical component in regulating cellular contraction, motility, and structural integrity. In normal physiology, MYL9 is particularly important for smooth muscle function, with MYL9 deficiency in mice causing lethal abnormalities in multiple organs including the bladder, small intestine, and lungs .
Selection of an appropriate MYL9 antibody requires careful consideration of several factors:
Specificity concerns: MYL9 shares >96% amino acid homology with MYL12A and MYL12B, leading to significant cross-reactivity of antibodies. Consider using genetic knockdown controls to validate specificity.
Application compatibility: Verify that the antibody has been validated for your specific application (Western blot, IHC, IF, ELISA).
Host species: Select an antibody raised in a species that complements your experimental system to avoid cross-reactivity in multi-antibody studies.
Epitope location: For phosphorylation studies, ensure the antibody recognizes the appropriate phosphorylated or non-phosphorylated form.
Tissue expression: MYL9 shows tissue-specific expression patterns. For instance, it's restricted to the muscularis propria of the small intestine and bladder, and in smooth muscle layers of bronchi and major vessels .
MYL9 antibodies are utilized across multiple research applications:
Western blotting: The most widely used application for detecting MYL9 protein expression levels in cell and tissue lysates.
Immunohistochemistry (IHC): Used to visualize MYL9 expression patterns in tissue sections, particularly valuable for studying smooth muscle and cancer tissue samples.
Immunofluorescence (IF): Enables subcellular localization studies of MYL9 protein.
ELISA: Quantitative measurement of MYL9 in biological samples.
Chromatin immunoprecipitation (ChIP): When studying transcriptional regulation involving MYL9.
Each application requires specific optimization parameters including antibody concentration, incubation time, and buffer conditions .
Validating MYL9 antibody specificity is particularly challenging due to high homology with related proteins. A comprehensive validation approach includes:
Genetic knockdown validation: Using siRNA targeting MYL9 (as demonstrated in research where siRNA-MYL9-1 and siRNA-MYL9-2 were employed to silence MYL9 expression) provides the most conclusive validation method .
Overexpression control: Testing antibody reactivity in systems with ectopic MYL9 overexpression (e.g., Ov-MYL9 transfected cells) .
Knockout tissues: If available, tissues from MYL9 knockout models serve as gold-standard negative controls. Studies using MYL9-deficient mice have shown the complete absence of antibody signal in tissues that normally express MYL9 .
Multiple antibody validation: Using different antibodies targeting distinct epitopes of MYL9.
Western blot molecular weight verification: Confirming reactivity at the expected 19.8 kDa range.
Cross-reactivity assessment: Evaluating potential cross-reactivity with MYL12A and MYL12B by comparing expression patterns with known tissue-specific distribution patterns .
Successful Western blotting for MYL9 requires careful optimization:
Protein extraction: Use RIPA lysis buffer containing phenylmethanesulfonyl fluoride and phosphatase inhibitors (at a ratio of 100:1:1) to effectively extract total protein while preserving phosphorylation states.
Protein loading: 30 μg of protein is typically sufficient for detection in cells with moderate MYL9 expression.
Gel percentage: 10% SDS-PAGE gels provide optimal separation for the 19.8 kDa MYL9 protein.
Transfer conditions: Standard PVDF membranes are suitable for MYL9 transfer.
Blocking: 5% bovine serum albumin in TBST for 1 hour at 37°C minimizes background.
Primary antibody: Incubate with anti-MYL9 antibody (1:1000 dilution is typically effective) overnight at 4°C.
Secondary antibody: HRP-conjugated anti-mouse or anti-rabbit IgG (depending on primary antibody host) at 37°C for 1 hour.
Detection: Standard ECL detection systems are sufficient for visualizing MYL9 bands.
Include appropriate loading controls such as β-tubulin or GAPDH for normalization .
For comprehensive MYL9 gene expression analysis:
RT-qPCR protocol:
Extract total RNA using TRIzol reagent
Synthesize cDNA from 1 μg of total RNA using reverse transcriptase
Perform qPCR using SYBR Green with specific primers
Recommended primer sequences:
MYL9 forward: 5′-GCCACATCCAATGTCTTCGC-3′
MYL9 reverse: 5′-GCGTTGCGAATCACATCCTC-3′
Use GAPDH as internal control
Calculate relative expression using the 2^-ΔΔCt method
Thermocycling conditions:
Initial denaturation at 95°C for 10 minutes
45 cycles of: 95°C for 15s, 60°C for 15s, and 72°C for 10s
Confirm specificity with melting curve analysis
Expression normalization: For comparative studies, set the expression in control samples as 1 (such as peritumoral tissues in cancer studies) .
MYL9 has been identified as a key regulator in colorectal cancer (CRC) progression through multiple mechanisms:
Overexpression pattern: MYL9 protein and mRNA expression is significantly upregulated in colorectal cancer cell lines (SW480, SW620, HT-29, and HCT116) compared to normal colorectal epithelial cells (NCM460) .
Functional impacts on cancer hallmarks:
Proliferation: MYL9 knockdown using siRNA significantly decreases CRC cell proliferation, while overexpression enhances it.
Migration and invasion: MYL9 silencing inhibits, while overexpression promotes, the migration and invasion capabilities of CRC cells.
Metastasis-related proteins: MYL9 modulates the expression of matrix metalloproteinases (MMP2 and MMP9), with knockdown decreasing and overexpression increasing these metastasis-promoting proteins .
Molecular mechanism: MYL9 promotes CRC progression by binding to YAP1 and thereby activating Hippo signaling, a pathway critical for tumor growth and metastasis .
Microenvironment regulation: Interestingly, MYL9 is predominantly expressed in cancer-associated fibroblasts (CAFs) rather than CRC cells themselves. In this context, MYL9 regulates the secretion of CCL2 and TGF-β1 from CAFs, which affects the immune microenvironment and CRC progression .
To effectively investigate MYL9's impact on cancer cell migration and invasion:
Genetic manipulation approaches:
Migration assays:
Invasion assays:
Co-culture systems for stromal interaction studies:
Colony formation assay:
MYL9 has been discovered to significantly impact cancer cell metabolism, particularly aerobic glycolysis (the Warburg effect):
Glycolytic enhancement: Research has demonstrated that MYL9 promotes squamous cervical cancer (SCC) migration and invasion specifically by enhancing aerobic glycolysis .
Molecular mechanism: MYL9 increases the activity of the JAK2/STAT3 signaling pathway, which is known to regulate metabolic reprogramming in cancer cells .
Glycolytic markers regulated by MYL9:
GLUT1 (glucose transporter 1): Facilitates increased glucose uptake
HK2 (hexokinase 2): Catalyzes the first rate-limiting step of glycolysis
LDHA (lactate dehydrogenase A): Converts pyruvate to lactate, enabling continued glycolytic flux
Measurement approaches:
Therapeutic implications: The connection between MYL9 and glycolysis suggests that targeting MYL9 could potentially disrupt cancer metabolism, offering a novel therapeutic strategy.
Addressing MYL9 antibody cross-reactivity requires sophisticated experimental design:
Tissue selection strategy: Leverage differential expression patterns. For instance, bladder and intestine tissues express high levels of MYL9 but low levels of MYL12A and MYL12B, making them suitable for MYL9-focused studies .
Reporter gene systems: Consider using genetic models with reporter genes like the LacZ-knockin/knockout mouse model, which allows precise mapping of MYL9 expression without relying solely on antibody detection .
RNA-based validation: Complement protein studies with RNA analysis techniques (RT-qPCR or RNA-seq) using MYL9-specific primers to verify expression patterns .
Bioinformatic verification: Compare your findings with tissue expression databases (such as GTEx Portal) to confirm expected distribution patterns for MYL9 versus MYL12A/B .
Genetic knockout controls: The most definitive approach is using tissue from MYL9 knockout models as negative controls, as demonstrated in studies where antibody staining was detected in wild-type tissues but absent in MYL9-deficient tissues .
Research has revealed distinct roles for MYL9 depending on its cellular context:
Cell-specific expression patterns:
Indirect versus direct effects:
Methodological considerations:
For tumor microenvironment studies: Co-culture systems, conditioned media experiments, and in vivo models are essential.
For direct cellular function: Standard cell-autonomous assays (proliferation, migration) with genetic manipulation are appropriate.
Therapeutic targeting implications:
Emerging research suggests important connections between MYL9 and anti-tumor immunity:
Immunosuppressive microenvironment: High MYL9 expression in CAFs is associated with M2 macrophage infiltration, creating an immunosuppressive microenvironment in colorectal cancer that reduces sensitivity to immunotherapy .
Integration methodologies:
Analyze MYL9 expression in relation to immune cell infiltration markers
Assess correlation between MYL9 levels and response to immunotherapy in patient cohorts
Design combination studies targeting both MYL9 and immune checkpoints
Experimental approaches:
Flow cytometry to characterize immune infiltrates in MYL9-high versus MYL9-low tumors
Single-cell RNA sequencing to map MYL9 expression across tumor and immune cell populations
Multiplex immunohistochemistry to visualize spatial relationships between MYL9-expressing cells and immune cells
Mechanistic investigations: Explore how MYL9-regulated factors (like CCL2 and TGF-β1) modulate T cell function, immune checkpoint expression, and antigen presentation .
When confronting weak or absent MYL9 signals in Western blots:
Sample preparation optimization:
Ensure complete protein extraction using RIPA buffer with protease inhibitors
Avoid repeated freeze-thaw cycles of protein samples
Add phosphatase inhibitors if studying phosphorylated MYL9
Antibody condition assessment:
Verify antibody storage conditions and expiration date
Titrate antibody concentration (try ranges from 1:500 to 1:2000)
Extend primary antibody incubation (overnight at 4°C is optimal)
Expression verification:
Technical optimization:
Increase protein loading (up to 50 μg)
Use more sensitive detection systems (enhanced chemiluminescence plus)
Optimize transfer conditions (lower voltage for longer time for small proteins)
When facing discrepancies between protein and RNA detection methods:
Validation approaches:
Perform experiments with multiple antibodies targeting different MYL9 epitopes
Use genetic manipulation (siRNA, CRISPR) to confirm specificity
Employ alternative protein detection methods (mass spectrometry)
Biological explanations to consider:
Post-transcriptional regulation: MYL9 mRNA can be actively degraded by mechanisms like DROSHA-mediated decay, resulting in mRNA expression without corresponding protein
Protein stability differences: Variations in protein half-life can cause disconnects between mRNA and protein levels
Cell-type heterogeneity: In tissues with mixed cell populations, bulk RNA methods may detect signal from minority populations
Resolution strategies:
Single-cell approaches to resolve cellular heterogeneity
Polysome profiling to assess translation efficiency
Protein degradation studies using proteasome inhibitors
Documentation best practices: Clearly report and discuss discrepancies in publications rather than selectively reporting concordant results .
MYL9 shows significant potential as a cancer biomarker:
MYL9 has been linked to epithelial-mesenchymal transition (EMT), a critical process in cancer progression:
Experimental design considerations:
Monitor classical EMT markers (E-cadherin, vimentin, Snail, Slug) alongside MYL9 manipulation
Use both 2D and 3D culture systems to capture different aspects of EMT
Employ live-cell imaging to observe morphological changes in real-time
Mechanistic investigations:
Explore connections between MYL9 and TGF-β signaling, as MYL9 regulates TGF-β1 secretion from CAFs
Investigate cytoskeletal remodeling, as MYL9's role in contractile function may directly impact cell shape changes during EMT
Study potential interactions with IQGAP1, which binds MYL9 and regulates cytoskeletal dynamics
In vivo approaches:
Develop metastasis models with MYL9 manipulation
Analyze circulating tumor cells for MYL9 expression
Employ lineage tracing to follow EMT processes in MYL9-altered settings
Translational relevance: