MYL12A’s activity is modulated by two kinase systems:
Myosin Light Chain Kinase (MLCK): Phosphorylates MYL12A at specific residues, enhancing myosin II ATPase activity and actin binding .
Rho-Kinase (ROCK): Indirectly regulates MYL12A via inhibition of myosin phosphatase, prolonging phosphorylation states .
MYL12A acts as a ligand for CD69, a receptor expressed on activated T cells. This interaction facilitates leukocyte recruitment to inflamed tissues, particularly in airway inflammation and eosinophilic chronic rhinosinusitis .
MYL12A is implicated in several pathologies:
MYL12A expression is influenced by environmental and therapeutic agents:
Chemical | Effect | Reference |
---|---|---|
Benzo[a]pyrene | Increases mRNA expression . | |
Bisphenol A | Conflicting reports (↑/↓ expression) . | |
Methotrexate | Elevates mRNA levels . | |
Doxorubicin | Alters protein expression . |
MYL12A intersects with key signaling networks:
Rho GTPase Pathways: Regulates actin cytoskeleton remodeling .
MAPK Pathways: MYL12B (paralog) is a substrate for MAPK kinases, indirectly linking to MYL12A function .
Myosin regulatory light chain 12A, MLC-2B, Myosin RLC, Myosin regulatory light chain 2 nonsarcomeric, Myosin regulatory light chain MRLC3, MYL12A, MLCB, MRLC3, RLC, MRCL3, MYL2B.
MGSSHHHHHH SSGLVPRGSH MGSHMSSKRT KTKTKKRPQR ATSNVFAMFD QSQIQEFKEA FNMIDQNRDG FIDKEDLHDM LASLGKNPTD EYLDAMMNEA PGPINFTMFL TMFGEKLNGT DPEDVIRNAF ACFDEEATGT IQEDYLRELL TTMGDRFTDE EVDELYREAP IDKKGNFNYI EFTRILKHGA KDKDD.
MYL12A is a non-sarcomeric myosin regulatory light chain (MRLC) that belongs to the class II MRLCs present in non-muscle cells. It functions as a regulatory component of the myosin II complex, which consists of myosin heavy chains (MHCs), regulatory light chains (RLCs), and essential light chains (ELCs) . MYL12A acts as a critical myosin regulatory subunit that controls contractile activity in both smooth muscle and non-muscle cells through phosphorylation mechanisms .
The primary functions of MYL12A include:
Regulation of cellular morphology and dynamics
Control of cytoskeletal organization and stability
Modulation of cell adhesion, migration, and division
Contribution to cytokinesis, receptor capping, and cell locomotion
Potential involvement in DNA damage repair by sequestering the transcriptional regulator apoptosis-antagonizing transcription factor (AATF)/Che-1
Methodologically, when studying MYL12A's basic functions, researchers typically employ immunofluorescence staining to visualize its cellular localization, often finding it expressed in the apical portion of specific cell types such as hair cells in auditory epithelia .
MYL12A shares high sequence homology with two other non-muscle myosin regulatory light chains, MYL12B and MYL9 . While functionally similar, there are several distinguishing features:
Classification: MYL12A belongs to class II MRLCs present in non-muscle cells, distinct from class I MRLCs found in muscles (such as MLC1, MLC2, MLC3, MYL4, MYL5, MYL6, MYL7)
Phosphorylation sites: Class II MRLCs like MYL12A are phosphorylated mainly at Ser19, while class I MRLCs are phosphorylated at Ser17
Expression patterns: Proteomic analysis has shown that despite their similarities, MYL12A, MYL12B, and MYL9 may have distinct tissue-specific expression profiles
Protein interactions: MYL12A associates with specific myosin heavy chains, particularly MYH9 (NMHC IIA) and MYH10 (NMHC IIB), as well as the essential light chain MYL6 in fibroblasts
For experimental differentiation between these highly homologous proteins, researchers typically rely on specific antibodies or gene-specific probes for droplet digital PCR, as demonstrated in studies of auditory epithelial cells .
Studying MYL12A phosphorylation states requires multiple complementary approaches:
Antibody-based detection methods:
Immunoblotting with phospho-specific antibodies such as anti-phospho-myosin light chain 2 (Cell Signaling Technology, #3671) at 1:50 dilution for immunostaining
Immunofluorescence microscopy to visualize cellular distribution of phosphorylated MYL12A
Flow cytometry for quantitative assessment of phosphorylation levels
Protein purification and in vitro analysis:
Expression of recombinant MYL12A in bacterial systems using Gateway Technology with specific primers (forward: 5′-caccatgtcgagcaaaaaggcaaagaccaa-3′; reverse: 5′-tcagtcatctttgtctttggctccatgttt-3′)
Purification using immobilized metal ion affinity chromatography (IMAC) with TALON Metal Affinity Resin
In vitro phosphorylation assays with purified kinases such as smooth muscle MLCK (smMLCK)
Pharmacological approaches:
Application of kinase inhibitors like ML-7 (smMLCK inhibitor) to study the effects of reduced phosphorylation
Comparison of cell area expansion or morphological changes before and after inhibitor treatment
Mass spectrometry:
Phosphopeptide enrichment followed by LC-MS/MS to identify specific phosphorylation sites and quantify phosphorylation levels
These methodological approaches allow researchers to correlate phosphorylation status with functional outcomes, as demonstrated in studies showing that MYL12 phosphorylation by smMLCK contributes to apical constriction-like cellular shape changes .
An optimal experimental design for investigating MYL12A's role in cell morphology should include:
Knockdown/knockout approaches:
siRNA or shRNA-mediated knockdown of MYL12A in cell culture models
CRISPR-Cas9-mediated gene editing for complete knockout
Analysis of resulting morphological changes using time-lapse microscopy and quantitative image analysis
Research has shown that knockdown of MYL12A/12B in NIH 3T3 fibroblasts results in striking changes in cell morphology and dynamics, with significant reductions in the levels of associated proteins including MYH9, MYH10, and MYL6 .
Rescue experiments:
Re-introduction of wild-type MYL12A after knockdown
Expression of phosphomimetic (S19D) or non-phosphorylatable (S19A) mutants to assess the role of phosphorylation
Comparative analysis of morphological rescue efficiency
Live cell imaging techniques:
Fluorescently tagged MYL12A constructs for real-time visualization
Quantification of cell area, shape, protrusion formation, and migration parameters
Correlation of morphological changes with MYL12A localization and dynamics
Cytoskeletal co-localization studies:
Immunofluorescence staining of MYL12A (using antibodies such as anti-MYL12A/B at 1:50 dilution) alongside actin and other cytoskeletal components
Super-resolution microscopy to resolve fine structural details
Analysis of stress fiber organization and focal adhesion formation
Pharmacological interventions:
Application of cytoskeletal disruptors or stabilizers
Use of kinase inhibitors to modulate MYL12A phosphorylation
Correlation between phosphorylation status and morphological outcomes
This comprehensive experimental approach enables researchers to establish causal relationships between MYL12A function and specific aspects of cellular morphology.
MYL12A forms critical interactions with myosin heavy chains that are essential for the structural integrity and function of the myosin II complex. These interactions can be characterized through several methodological approaches:
Proteomic analysis of protein complexes:
Immunoprecipitation followed by mass spectrometry has revealed that MYL12A associates with specific myosin heavy chains, particularly MYH9 (NMHC IIA) and MYH10 (NMHC IIB), as well as the essential light chain MYL6 in NIH 3T3 fibroblasts
Blue native gel electrophoresis to assess intact complexes
Interaction mapping:
Yeast two-hybrid screening to identify specific binding domains
In vitro binding assays with purified components
Truncation mutants to define interaction domains
Structural biology approaches:
X-ray crystallography or cryo-electron microscopy of the assembled complex
Molecular dynamics simulations to predict interaction interfaces
Hydrogen-deuterium exchange mass spectrometry to map interaction surfaces
Functional consequence analysis:
Knockdown studies have demonstrated that reduced levels of MYL12A/12B lead to significant reductions in the levels of associated proteins including MYH9, MYH10, and MYL6, indicating that these regulatory light chains are essential for maintaining the stability of the entire myosin II complex
Expression of interaction-deficient mutants and assessment of complex formation
Cross-linking and proximity labeling:
Chemical cross-linking followed by mass spectrometry to identify interacting regions
BioID or APEX2 proximity labeling to identify the interactome in living cells
Comprehensive interaction analysis has disclosed that MYL12A can interact with a variety of MHC IIs in diverse cell and tissue types, but does so optimally with non-muscle types of MHC II . These findings provide direct evidence that normal levels of non-muscle regulatory light chains are essential for maintaining the integrity of myosin II.
The functional relationship between MYL12A, MYL12B, and MYL9 in human cells is characterized by both overlapping and distinct roles:
Sequence and structural similarities:
These three proteins share high sequence homology, suggesting evolutionary conservation of critical functions
All three belong to class II myosin regulatory light chains present in non-muscle cells
Expression patterns:
Droplet digital PCR analysis has demonstrated that different cell types may express varying levels of these three proteins
For example, in auditory hair cells (HCs), both MYL12A and MYL12B are expressed, while MYL9 expression is observed primarily in inner sulcus cells (ISCs)
Subcellular localization differences:
Immunofluorescence staining has shown that MYL12 (A/B) is expressed in the apical portion of hair cells, whereas both MYL12 and MYL9 are expressed on inner sulcus cells
These localization differences suggest specialized functions in different cellular compartments
Functional redundancy and specificity:
Co-regulation and compensation:
Studies suggest potential compensatory mechanisms, where deficiency in one protein may be partially compensated by the others
This is evidenced by the fact that knockdown of both MYL12A and MYL12B is often necessary to observe dramatic phenotypic changes
Methodology for studying their relationships:
Simultaneous or sequential knockdown/knockout of each gene
Rescue experiments with each protein to assess functional equivalence
Comparative phosphoproteomics to identify regulatory differences
Protein-protein interaction mapping to define unique binding partners
Understanding the complex relationship between these three highly related proteins requires integrated approaches that can distinguish their specific functions while accounting for potential redundancy and compensation.
MYL12A phosphorylation serves as a molecular switch that regulates cell contractility and shape changes through several interconnected mechanisms:
Biochemical activation of myosin motor activity:
Phosphorylation of MYL12A, primarily at Ser19, increases the Mg²⁺-ATPase activity of myosin heavy chains
This enhanced ATPase activity enables increased cross-bridge cycling and contractile force generation
In vitro studies have confirmed that purified MYL12B (highly homologous to MYL12A) can be phosphorylated by smooth muscle myosin light chain kinase (smMLCK)
Cytoskeletal reorganization:
Phosphorylated MYL12A promotes assembly of non-muscle myosin II filaments
These assembled filaments interact with actin to form contractile actomyosin networks
The resulting contractile forces drive cellular shape changes, such as apical constriction in epithelial cells
Experimental evidence in cellular models:
In hair cells of auditory epithelia, inhibition of smMLCK with ML-7 leads to reduced MYL12 phosphorylation, accompanied by expansion of the cell area of outer hair cells
This observation directly links MYL12 phosphorylation to the maintenance of apical constriction-like cellular shape
Knockdown studies in NIH 3T3 fibroblasts have demonstrated that MYL12A/B are critical for cell structure and dynamics
Regulatory pathway integration:
MYL12A phosphorylation serves as a convergence point for multiple signaling pathways
Kinases such as MLCK, ROCK, MRCK, and Citron kinase can phosphorylate MYL12A
Phosphatases like myosin phosphatase (MYPT1-PP1) counterbalance kinase activity
This allows for precise spatial and temporal control of cellular contractility
Methodology for experimental investigation:
Phospho-specific antibodies to detect MYL12A phosphorylation states (e.g., anti-phospho-myosin light chain 2, Cell Signaling Technology, #3671)
Kinase inhibitors like ML-7 to modulate phosphorylation levels
Live cell imaging with fluorescent reporters to visualize dynamic shape changes
Traction force microscopy to measure contractile forces generated by cells
These findings collectively demonstrate that MYL12A phosphorylation by kinases such as smMLCK contributes significantly to the regulation of cell shape through modulation of actomyosin contractility.
MYL12A plays a crucial role in cytokinesis and cell division through its regulation of contractile ring formation and function:
Contractile ring assembly and function:
As a regulatory component of non-muscle myosin II, phosphorylated MYL12A contributes to the assembly and contraction of the actomyosin ring during cytokinesis
The spatiotemporal regulation of MYL12A phosphorylation helps define the cleavage furrow
MYL12A's interaction with myosin heavy chains MYH9 and MYH10 is critical for proper contractile ring function
Mechanistic involvement:
During cell division, MYL12A localizes to the cleavage furrow
Its phosphorylation increases myosin II ATPase activity and promotes interaction with actin filaments
This activation generates the contractile force necessary for furrow ingression and eventual daughter cell separation
Experimental approaches to study MYL12A in cytokinesis:
Live cell imaging with fluorescently tagged MYL12A to track its dynamics during cell division
Phospho-specific antibodies to monitor phosphorylation state changes throughout the cell cycle
Small molecule inhibitors of kinases/phosphatases that regulate MYL12A to assess functional consequences
RNAi or CRISPR-based depletion followed by detailed phenotypic analysis of division defects
Phenotypic consequences of disruption:
Knockdown of MYL12A/12B in fibroblasts causes striking changes in cell morphology and dynamics
Cytokinesis failures can lead to binucleation or polyploidy
Abnormal MYL12A function may contribute to genomic instability in pathological contexts
Integration with regulatory pathways:
Cell cycle-dependent kinases indirectly control MYL12A phosphorylation state
RhoA-ROCK signaling is a major regulatory pathway for MYL12A phosphorylation during cytokinesis
Aurora B kinase may contribute to MYL12A regulation at the midbody
These mechanisms highlight MYL12A's central importance in orchestrating the mechanical aspects of cell division, with significant implications for understanding both normal cellular function and pathological states associated with division defects.
MYL12A has been implicated in several human disease processes through its central role in cellular contractility, morphology, and division:
Disease associations:
Cancer progression: Altered MYL12A expression or phosphorylation has been linked to cancer cell migration, invasion, and metastasis
Cardiovascular disorders: MYL12A phosphorylation levels have been associated with myocardial injury and hypertrophy
Orbital Granuloma and Hemoglobin E Disease: GeneCards indicates associations between MYL12A and these specific conditions
Methodological approaches for studying disease associations:
Expression analysis in patient samples:
Quantitative PCR to measure mRNA levels
Immunohistochemistry to assess protein expression and localization in diseased tissues
Western blotting with phospho-specific antibodies to evaluate activation state
Functional studies in disease models:
CRISPR-Cas9 editing to recreate disease-associated mutations
Patient-derived cells to study phenotypic consequences
In vitro disease models using relevant cell types
Chemical interaction profiling:
Studies have shown that MYL12A expression or function can be affected by various chemicals, including:
Carbon tetrachloride (increases MYL12A mRNA expression)
Folic acid (increases MYL12A mRNA expression when co-treated with 1,2-dimethylhydrazine)
17β-estradiol (can decrease MYL12A mRNA expression in certain contexts)
Tetrachlorodibenzodioxin (affects MYL12A expression and promotes binding of AHR protein to MYL12A promoter)
Genetic association studies:
Genome-wide association studies to identify MYL12A variants linked to disease risk
Targeted sequencing of the MYL12A gene in patient cohorts
Expression quantitative trait loci (eQTL) analysis
Pathway analysis:
Integration with known disease pathways, such as:
Clinical correlation studies:
Association of MYL12A expression or phosphorylation with clinical outcomes
Potential biomarker development based on MYL12A status
These methodological approaches provide a comprehensive framework for investigating the complex roles of MYL12A in human disease processes, potentially leading to new diagnostic or therapeutic strategies.
While MYL12A is emerging as a potential therapeutic target, research in this area is still developing. Current and emerging strategies include:
Kinase inhibition approaches:
Targeting the kinases that phosphorylate MYL12A, such as smooth muscle myosin light chain kinase (smMLCK)
ML-7, an inhibitor of smMLCK, has been shown to reduce MYL12 phosphorylation and affect cell morphology in experimental models
ROCK inhibitors like Y-27632 that indirectly affect MYL12A phosphorylation status
Gene expression modulation:
RNA interference approaches to modulate MYL12A levels in pathological contexts
Antisense oligonucleotides targeting MYL12A mRNA
CRISPR-based technologies for precise genomic editing
Protein-protein interaction disruptors:
Small molecules or peptides designed to disrupt specific interactions between MYL12A and myosin heavy chains
This approach is supported by evidence that MYL12A interactions with MYH9 and MYH10 are critical for myosin II complex stability
Phosphorylation site-specific approaches:
Development of phosphorylation site-specific modulators
Phosphomimetic peptides that compete with endogenous MYL12A
Methodological considerations for therapeutic development:
High-throughput screening assays using recombinant MYL12A protein
Cellular assays monitoring phosphorylation status and contractility
In vitro reconstitution of myosin II complexes to screen for stabilizers or disruptors
Structure-based drug design targeting MYL12A or its interaction interfaces
Potential applications in disease contexts:
Cancer: Targeting the migratory and invasive capabilities of tumor cells by modulating MYL12A function
Cardiovascular disorders: Addressing pathological hypertrophy by normalizing MYL12A phosphorylation levels
Fibrotic conditions: Reducing excessive contractility in fibrotic tissues
Chemical agents affecting MYL12A:
Several compounds have been identified that affect MYL12A expression, including bisphenol S (increases MYL12A mRNA expression)
These findings may provide starting points for therapeutic development
These emerging strategies highlight the potential of MYL12A as a therapeutic target, though significant research is still needed to translate these approaches into clinical applications.
MYL12A expression and regulation exhibit tissue-specific patterns and dynamic changes in disease states:
Tissue-specific expression patterns:
Droplet digital PCR and immunofluorescence staining have revealed differential expression of MYL12A across tissues
In auditory epithelia, MYL12A/B is expressed in hair cells, while both MYL12A/B and MYL9 are expressed in inner sulcus cells
Expression can be localized to specific subcellular regions, such as the apical portion of hair cells
Transcriptional regulation:
Promoter analysis has identified binding sites for transcription factors
AHR (aryl hydrocarbon receptor) has been shown to bind to the MYL12A promoter after tetrachlorodibenzodioxin treatment
Different tissue types likely employ distinct transcriptional programs to regulate MYL12A expression
Hormonal regulation:
Evidence suggests hormonal influence on MYL12A expression:
Disease state alterations:
Cancer: Altered expression patterns in various tumor types
Cardiovascular disorders: Changes in MYL12A phosphorylation associated with myocardial injury and hypertrophy
Other conditions: Association with Orbital Granuloma and Hemoglobin E Disease
Environmental and chemical influences:
MYL12A expression responds to various chemical exposures:
Methodological approaches for studying expression patterns:
Single-cell RNA sequencing to characterize cell-type-specific expression
Tissue microarrays for protein-level assessment across multiple samples
Chromatin immunoprecipitation sequencing (ChIP-seq) to identify regulatory elements
ATAC-seq to assess chromatin accessibility at the MYL12A locus
Reporter assays to study promoter activity in different cellular contexts
These varied expression patterns and regulatory mechanisms highlight the complex contextual control of MYL12A, which likely contributes to its diverse roles in different tissues and its involvement in various pathological processes.
While phosphorylation is the most well-characterized post-translational modification (PTM) of MYL12A, research suggests that other PTMs may also regulate its function:
Types of potential post-translational modifications:
Acetylation:
May affect protein-protein interactions
Could influence MYL12A's binding affinity for myosin heavy chains
Potentially regulates protein stability
Ubiquitination:
Likely regulates MYL12A protein turnover and degradation
May be involved in quality control of myosin II complex assembly
Could be dynamically regulated during cell cycle progression
Methylation:
Potentially modulates interaction with binding partners
May affect nuclear localization or other subcellular targeting
SUMOylation:
Could alter protein-protein interaction networks
May regulate stability or activity in specific cellular compartments
Methodological approaches for studying PTMs:
Mass spectrometry-based proteomics:
Shotgun proteomics to identify novel PTMs
Targeted MS approaches to quantify specific modifications
Enrichment strategies for specific PTM types prior to MS analysis
Site-specific mutants:
Generation of lysine-to-arginine mutants to prevent acetylation/ubiquitination
Expression of mutant proteins in knockout backgrounds to assess functional consequences
PTM-specific antibodies:
Development and validation of antibodies against specific modified forms
Immunoprecipitation combined with western blotting to detect modified populations
Crosstalk with phosphorylation:
Investigation of how other PTMs may enhance or inhibit phosphorylation at Ser19
Assessment of how multiple PTMs may combine to create a complex regulatory code
Functional consequences:
Effects on myosin II complex stability and integrity
Alterations in contractile properties or force generation
Changes in interaction with regulatory proteins or cytoskeletal components
Potential influence on MYL12A's reported role in DNA damage repair through sequestration of the transcriptional regulator AATF/Che-1
While direct evidence for many of these modifications in MYL12A is still emerging, these represent important directions for future research to fully understand the complex regulation of this critical cytoskeletal regulatory protein.
MYL12A demonstrates significant evolutionary conservation across species, providing valuable insights into its fundamental biological importance:
Sequence conservation:
High sequence homology exists between human MYL12A and its orthologs in other mammals
The functional domains, particularly those involved in calcium binding and phosphorylation sites, show strong conservation
This conservation extends to non-mammalian vertebrates, indicating ancient evolutionary origins
Comparative experimental approaches:
Mouse models are frequently used to study MYL12A function due to the high conservation between mouse and human proteins
Evidence from citations indicates that findings in mouse models are often considered relevant to human MYL12A function
Experimental tools designed for one species often cross-react with orthologs from related species due to high sequence similarity
Evolutionary insights:
The presence of multiple related isoforms (MYL12A, MYL12B, MYL9) suggests gene duplication events during evolution
These paralogs likely evolved specialized functions while maintaining core regulatory capabilities
The high conservation of MYL12A suggests strong evolutionary pressure against sequence variation, highlighting its essential cellular functions
Methodological considerations for evolutionary studies:
Phylogenetic analysis to trace the evolutionary history of MYL12A and related proteins
Comparative genomics to identify conserved regulatory elements across species
Experimental validation in model organisms to test functional conservation
Translational implications:
The high conservation suggests that findings from model organisms likely translate well to human biology
Understanding species-specific differences may help explain unique aspects of human cytoskeletal regulation
Evolutionary conservation supports the use of diverse model systems for studying MYL12A function
Researchers interested in evolutionary aspects should consider employing comparative genomics, functional assays across multiple species, and phylogenetic analyses to gain deeper insights into the evolutionary trajectory and functional significance of MYL12A conservation.
Comparative analysis of MYL12A with other myosin regulatory light chains across biological systems reveals important insights into functional specialization and regulatory diversity:
Structural and functional comparisons:
MYL12A shares high sequence homology with MYL12B and MYL9, but has distinct roles from muscle-specific regulatory light chains like MLC1, MLC2, and MLC3
Class I MRLCs (muscle-specific) and class II MRLCs (non-muscle, including MYL12A) differ in their phosphorylation sites: Ser17 for class I versus Ser19 for class II
These differences reflect evolutionary adaptations to different physiological demands in muscle versus non-muscle tissues
Comparative expression patterns:
Droplet digital PCR analysis has revealed tissue-specific expression patterns of various MRLCs
In auditory epithelia, differential expression of MYL12A/B and MYL9 corresponds to specific cellular locations and functions
Understanding these expression patterns helps elucidate why multiple, highly similar proteins are maintained in the genome
Methodological approaches for comparative studies:
Parallel knockdown/knockout experiments of different MRLCs to assess functional redundancy or specialization
Chimeric protein constructs to identify domain-specific functions
Cross-species complementation assays to test functional conservation
Regulatory mechanism diversity:
Different MRLCs may be regulated by distinct kinase and phosphatase systems
MYL12A is phosphorylated by smooth muscle MLCK (smMLCK) in certain cellular contexts
Comparative study of regulatory mechanisms reveals how different cellular systems have evolved specialized control mechanisms
Applications in biotechnology and medicine:
Understanding the unique properties of each MRLC informs the design of specific inhibitors or modulators
Insights from comparative analysis may guide the development of tissue-specific or context-specific interventions
Identification of unique features may help design more specific experimental tools and reagents
This comparative approach provides a broader evolutionary and functional context for understanding MYL12A, revealing how similar proteins can evolve specialized functions while maintaining core regulatory capabilities in different biological systems.
Several critical unresolved questions about MYL12A represent promising avenues for future research:
Structural biology and interaction dynamics:
What is the detailed structural basis for MYL12A's interactions with different myosin heavy chains?
How do conformational changes upon phosphorylation alter these interactions?
Can structural biology approaches reveal novel druggable pockets or interfaces?
Regulatory networks and signaling integration:
How is MYL12A regulation integrated with other cytoskeletal regulatory pathways?
What is the complete set of kinases and phosphatases that regulate MYL12A in different cellular contexts?
How do multiple signaling pathways converge to control MYL12A phosphorylation with spatial and temporal precision?
Novel functions beyond contractility:
What is the mechanistic basis for MYL12A's potential role in DNA damage repair through interaction with AATF/Che-1 ?
Does MYL12A participate in mechanotransduction or nuclear mechanobiology?
Are there additional, undiscovered functions in specialized cell types?
Disease mechanisms:
What are the precise mechanisms by which altered MYL12A function contributes to diseases like cancer and cardiovascular disorders ?
Are there specific MYL12A mutations associated with human disease phenotypes?
Could MYL12A serve as a prognostic biomarker or therapeutic target in specific pathological contexts?
Regulatory light chain specificity:
Why do cells express multiple, highly similar regulatory light chains (MYL12A, MYL12B, MYL9)?
What determines which regulatory light chain associates with specific myosin heavy chains in different contexts?
How is the balance between these similar proteins maintained and regulated?
Post-translational modification crosstalk:
How do different post-translational modifications beyond phosphorylation interact to regulate MYL12A?
Is there a "PTM code" that integrates multiple modifications to fine-tune function?
Methodological needs for addressing these questions:
Development of more specific antibodies and probes to distinguish between highly similar regulatory light chains
Advanced imaging approaches to visualize MYL12A dynamics in living cells with high spatial and temporal resolution
Precise genome editing tools to introduce specific mutations or tags at endogenous loci
Quantitative proteomics approaches to comprehensively map the MYL12A interactome and modification landscape
Addressing these questions will require interdisciplinary approaches combining structural biology, cell biology, biochemistry, and advanced imaging techniques, potentially yielding significant insights into both basic cytoskeletal biology and disease mechanisms.
Several cutting-edge technologies and approaches are poised to transform our understanding of MYL12A function:
Advanced imaging technologies:
Super-resolution microscopy: Techniques like STORM, PALM, or STED can resolve MYL12A localization at nanometer scales, revealing previously undetectable organization patterns
Lattice light-sheet microscopy: Enables long-term 3D imaging of MYL12A dynamics in living cells with minimal phototoxicity
Förster resonance energy transfer (FRET)-based sensors: Development of biosensors to visualize MYL12A phosphorylation or conformational changes in real time
Proteomics and interactomics approaches:
Proximity labeling methods: BioID or APEX2-based approaches to map the complete MYL12A interactome in living cells
Crosslinking mass spectrometry: Identifying direct interaction interfaces at amino acid resolution
Targeted proteomics: Precise quantification of MYL12A modifications and interactions across different conditions
Genomic and transcriptomic technologies:
Single-cell multi-omics: Integrated analysis of gene expression, chromatin accessibility, and protein levels in individual cells
Spatial transcriptomics: Mapping MYL12A expression patterns with spatial context in tissues
CRISPR screening: Genome-wide screens to identify novel regulators of MYL12A function
Structural biology innovations:
Cryo-electron microscopy: Determining high-resolution structures of MYL12A in complex with myosin heavy chains
Integrative structural biology: Combining multiple structural techniques to build comprehensive models of the myosin II complex
Single-molecule methods: Directly observing MYL12A-mediated conformational changes and force generation
Biophysical approaches:
Optical tweezers and magnetic tweezers: Measuring forces generated by MYL12A-containing actomyosin complexes at the single-molecule level
Traction force microscopy: Correlating cellular force generation with MYL12A activity
Micropattern and microfluidic systems: Controlled environments to study MYL12A function under defined geometric constraints
Systems biology and computational approaches:
Mathematical modeling: Integrating experimental data into predictive models of MYL12A regulation and function
Machine learning: Analyzing complex datasets to identify patterns and make predictions about MYL12A behavior
Network analysis: Placing MYL12A within the broader context of cellular signaling and regulatory networks
Organoid and tissue engineering technologies:
3D organoid cultures: Studying MYL12A function in more physiologically relevant contexts
Organ-on-chip platforms: Investigating MYL12A roles under controlled mechanical and biochemical conditions
Engineered tissues: Examining the contribution of MYL12A to tissue-level mechanics and organization
The integration of these emerging technologies promises to provide unprecedented insights into MYL12A's multifaceted roles in cellular function and disease, potentially revealing novel therapeutic opportunities and fundamental biological principles.
MYL12A encodes a nonsarcomeric myosin regulatory light chain. This protein is activated by phosphorylation and is involved in various cellular processes, including smooth muscle contraction, non-muscle cell contraction, cytokinesis, receptor capping, and cell locomotion . The gene is located on chromosome 18 and has several aliases, including MRLC3, MLCB, and MYL2B .
The human recombinant MYL12A protein is produced in E. coli and consists of a single, non-glycosylated polypeptide chain containing 195 amino acids, with a molecular mass of approximately 22.4 kDa. It is fused to a 24 amino acid His-tag at the N-terminus and purified using proprietary chromatographic techniques .
MYL12A is implicated in the regulation of smooth muscle and non-muscle cell contractile activity via its phosphorylation. It is also involved in DNA damage repair by sequestering the transcriptional regulator apoptosis-antagonizing transcription factor (AATF)/Che-1, which functions as a repressor of p53-driven apoptosis .