Cofilin-2 (CFL2), encoded by the CFL2 gene on human chromosome 14q13.2, is a muscle-specific actin-binding protein critical for regulating cytoskeletal dynamics . It belongs to the actin-depolymerizing factor (ADF) family, which includes cofilin-1 (CFL1) and destrin (DSTN) .
CFL2 mutations are rare causes of myofibrillar myopathy and congenital muscle disorders .
Parameter | Specification | Source |
---|---|---|
Purity | >90% (SDS-PAGE) | |
Tag | N-terminal His-tag (20 a.a.) | |
Storage | -20°C (long-term), 4°C (short-term) | |
Formulation | 20 mM Tris-HCl (pH 8.0), 50 mM NaCl |
Quantification methods:
The Human Cofilin-2 ELISA Kit (HUEB1994) enables precise measurement of serum/plasma levels:
Parameter | Specification |
---|---|
Detection range | 0.312–20 ng/mL |
Sensitivity | 0.156 ng/mL |
Sample types | Serum, plasma, cell culture supernatant |
Assay type | Sandwich ELISA |
This kit is critical for studying CFL2’s role in cancer, neurodegeneration, and autoimmune diseases .
The human CFL2 gene is localized on chromosome 14 and exhibits high homology to murine CFL2. Research has identified differentially spliced CFL2 transcripts, specifically CFL2a and CFL2b, with distinct tissue expression patterns. CFL2b is expressed predominantly in human skeletal muscle and heart, while CFL2a expression is observed across various tissues . The gene encodes a skeletal muscle-specific isoform of cofilin that is localized to the thin filaments, where it regulates actin dynamics through interactions with tropomyosins .
CFL2 functions as an actin-modulating protein that binds and depolymerizes filamentous F-actin while inhibiting the polymerization of monomeric G-actin in a pH-dependent manner . As a member of the AC protein family that includes cofilin-1 (CFL1) and destrin (DSTN), CFL2 plays a critical role in actin filament turnover necessary for proper muscle function. The protein's activity is regulated through phosphorylation/dephosphorylation cycles, with the dephosphorylated form being the active state that promotes actin filament disassembly .
Human CFL2 exists in two primary isoforms resulting from alternative splicing: CFL2a and CFL2b. Research demonstrates that CFL2b shows tissue-specific expression predominantly in human skeletal muscle and cardiac tissue. In contrast, CFL2a displays a broader expression pattern across various tissue types . This differential expression suggests specialized roles for these isoforms in tissue-specific functions, particularly in contractile tissues where actin dynamics are essential for physiological function.
Mutations in the CFL2 gene are associated with nemaline myopathy, a congenital muscle disorder characterized by muscle weakness and the presence of rod-like structures (nemaline bodies) in muscle fibers . The pathophysiological mechanism involves deficiency of functional cofilin-2 resulting in reduced depolymerization of actin filaments. This dysfunction causes abnormal accumulation of actin in nemaline bodies, minicores, and possibly concentric laminated bodies . The impaired actin turnover disrupts normal muscle function and contractility, ultimately manifesting as progressive muscle weakness in affected individuals.
Studies using mouse models of mechanically induced muscle damage have revealed that CFL2 expression changes significantly during muscle regeneration. The dephosphorylated (active) form of CFL2 emerges as the major isoform during later stages of muscle regeneration . A similar pattern of predominant dephosphorylated CFL2 has been observed in chronically regenerating dystrophin-deficient muscles from Duchenne muscular dystrophy patients . These findings suggest that CFL2 may play a crucial role in actin remodeling during the regenerative process, potentially facilitating the reorganization of the cytoskeleton required for myoblast fusion and myofiber formation.
Experimental evidence indicates that CFL2 acts as a negative regulator of skeletal muscle satellite cell proliferation and can induce cell apoptosis . This function has important implications for muscle maintenance and regeneration, as satellite cells serve as the primary stem cell population responsible for postnatal muscle growth and repair. The regulatory effect of CFL2 on satellite cell dynamics may be particularly important during embryonic development when the proliferation of muscle fibers is established, and in adult muscle where satellite cells remain quiescent until activated by injury or damage .
For comprehensive analysis of CFL2 expression, researchers should employ a multi-faceted approach combining RNA sequencing and protein detection methods. Transcription-level analysis through RNA-seq can identify differentially expressed CFL2 transcripts (CFL2a and CFL2b) . For protein-level detection, immunohistochemistry or immunofluorescence with CFL2-specific antibodies can localize the protein within tissues, while Western blotting can quantify expression levels and identify phosphorylated versus dephosphorylated forms . Additionally, in situ hybridization may be useful for distinguishing between the expression patterns of the different CFL2 isoforms in heterogeneous tissue samples.
To study CFL2 phosphorylation dynamics, researchers should employ phosphorylation-specific antibodies capable of distinguishing between phosphorylated (inactive) and dephosphorylated (active) forms of CFL2 . Quantitative Western blotting with these antibodies allows determination of the relative abundance of each form. For temporal studies of phosphorylation changes, time course experiments with appropriate stimuli (such as mechanical stress or growth factors) can reveal regulatory mechanisms. Advanced techniques such as phosphoproteomics using mass spectrometry can identify specific phosphorylation sites and potentially discover novel regulatory modifications. Additionally, in vitro kinase and phosphatase assays can help identify the specific enzymes responsible for modulating CFL2 activity.
Several experimental models are valuable for CFL2 research. Cell culture systems using C2C12 myoblasts or primary satellite cells allow for manipulation of CFL2 expression through overexpression or knockdown approaches to study effects on proliferation, differentiation, and actin dynamics . For in vivo studies, mouse models with mechanically induced muscle damage provide insights into regeneration processes . Genetic mouse models with CFL2 mutations or conditional knockouts can elucidate developmental roles and pathological mechanisms. Patient-derived muscle biopsies from individuals with nemaline myopathy or Duchenne muscular dystrophy offer opportunities to study CFL2 in human pathological contexts . Additionally, newer approaches using induced pluripotent stem cells differentiated into myogenic lineages can bridge the gap between cell culture and human relevance.
Genome-wide DNA methylation studies have revealed that CFL2 is among the genes strongly correlated with methylation-caused expression switches during embryonic muscle development . Research combining whole-genome bisulfite sequencing (WGBS) and RNA sequencing of muscle tissues collected at different embryonic development points has identified CFL2 as a key gene whose expression is influenced by changes in DNA methylation patterns . For researchers investigating this relationship, integrated methylation and expression analyses are essential to understand how epigenetic modifications regulate CFL2 during critical developmental windows. Methodologically, this requires careful timing of tissue collection across developmental stages, isolation of high-quality DNA and RNA from the same samples, and sophisticated bioinformatic approaches to correlate methylation changes with expression patterns.
CFL2 functions within a complex network of actin-regulating proteins, and understanding its protein-protein interactions is crucial for deciphering its mechanisms of action. Protein-protein interaction (PPI) networks have shown that CFL2 interacts with multiple partners in the regulation of actin dynamics . To systematically investigate these interactions, researchers should employ complementary approaches including co-immunoprecipitation followed by mass spectrometry to identify novel binding partners, proximity ligation assays to verify interactions in situ, and FRET/BRET approaches for dynamic interaction studies in living cells. Yeast two-hybrid screens and bacterial expression systems can help map specific binding domains. Additionally, in silico molecular modeling can predict structural interactions that can then be validated experimentally through site-directed mutagenesis of predicted interface residues.
Researchers studying CFL2 across different model systems (e.g., cell lines, animal models, human tissues) may encounter seemingly contradictory results regarding its function. To reconcile such discrepancies, several methodological approaches should be considered. First, carefully control for the specific CFL2 isoform being studied, as CFL2a and CFL2b may have distinct functions in different contexts . Second, characterize the phosphorylation state of CFL2 in each experimental system, as this dramatically affects its activity . Third, consider species-specific differences in CFL2 function and regulation when comparing human and animal model data. Fourth, account for cell-type specific contexts, particularly when comparing immortalized cell lines with primary cells or tissues. Finally, implement standardized experimental conditions when possible across different model systems to minimize technical variability.
For sensitive and specific detection of CFL2 in research and clinical samples, ELISA-based approaches have proven effective. The double antibody-sandwich ELISA method allows for quantitative detection of CFL2 with high sensitivity (as low as 9.375 pg/ml) and a detection range of 15.625-1000 pg/ml . This approach is suitable for various sample types including serum, plasma, cell culture supernatant, and tissue lysates . For researchers developing detection protocols, optimization of antibody specificity is critical to avoid cross-reactivity with other cofilin family members such as CFL1. Additionally, appropriate sample preparation methods should be established for different tissue types, particularly for muscle samples where protein extraction efficiency may vary depending on the state of the tissue (normal vs. pathological).
Distinguishing between phosphorylated and dephosphorylated CFL2 is essential for understanding its activity state in biological samples. Methodologically, this can be accomplished through several approaches. Phosphorylation-specific antibodies used in Western blotting or immunoprecipitation can directly identify the phosphorylation state . Two-dimensional gel electrophoresis can separate the differently charged phosphorylated forms. Phos-tag SDS-PAGE provides enhanced separation of phosphorylated proteins without requiring phospho-specific antibodies. For more detailed analysis, mass spectrometry-based phosphoproteomics can identify specific phosphorylation sites and their relative abundance. Additionally, functional assays measuring actin-binding and depolymerization activities can indirectly assess the proportion of active (dephosphorylated) CFL2 in samples.
CFL2 operates within a complex network of cytoskeletal regulators that collectively maintain muscle homeostasis. As part of the AC group of proteins that includes cofilin-1 (CFL1) and destrin (DSTN), CFL2 contributes to the dynamic regulation of actin filaments . Research approaches to understand this integrated function should include comprehensive interactome analyses to map connections between CFL2 and other cytoskeletal regulators. Co-expression studies can identify synergistic or antagonistic relationships between CFL2 and other actin-binding proteins in muscle tissues. Functional genomics approaches using CRISPR/Cas9 to create single and combination knockouts can reveal redundancies and unique functions among cytoskeletal regulators. Additionally, systems biology approaches integrating transcriptomic, proteomic, and functional data can help construct models of how these regulators work in concert to maintain muscle structure and function.
CFL2 research has significant implications for therapeutic development, particularly for nemaline myopathy and other muscle disorders characterized by cytoskeletal abnormalities. Understanding the molecular mechanisms by which CFL2 mutations lead to disease phenotypes can identify potential intervention points. For researchers pursuing therapeutic development, several methodological approaches are valuable: high-throughput screening for compounds that can restore actin dynamics in CFL2-deficient cells; gene therapy approaches to deliver functional CFL2 to affected tissues; antisense oligonucleotides to modulate splicing in cases where specific CFL2 isoforms are affected; and CRISPR-based genome editing to correct pathogenic mutations. Additionally, investigating the signaling pathways that regulate CFL2 phosphorylation could identify targets for pharmacological intervention to modulate CFL2 activity in diseased muscle.
Cofilin-2, also known as CFL2, is a member of the cofilin family of actin-binding proteins. This family includes cofilin-1 (CFL1) and destrin (DSTN), all of which play crucial roles in the regulation of actin filament dynamics . Cofilin-2 is specifically expressed in skeletal muscle tissue and is essential for muscle function and development .
Cofilin-2 is an actin-modulating protein that binds to filamentous F-actin and depolymerizes it, inhibiting the polymerization of monomeric G-actin in a pH-dependent manner . This activity is crucial for the dynamic reorganization of the actin cytoskeleton, which is essential for various cellular processes, including cell motility, division, and muscle contraction .
Recombinant human cofilin-2 is typically produced in Escherichia coli (E. coli) expression systems . The recombinant protein is often tagged with a His-tag to facilitate purification and is lyophilized for storage and transport . The purity of the recombinant protein is usually greater than 90%, making it suitable for various research applications .
Mutations in the CFL2 gene are associated with nemaline myopathy, a muscle disorder characterized by muscle weakness and the presence of rod-like structures called nemaline bodies in muscle fibers . Deficiency of cofilin-2 can lead to reduced depolymerization of actin filaments, causing their accumulation and contributing to the pathology of the disease .