Actin-binding protein involved in cellular motility and morphology. It inhibits actin polymerization, likely by sequestering G-actin.
KEGG: xla:379995
UniGene: Xl.63680
Xenopus laevis Twinfilin-2-A belongs to the evolutionarily conserved twinfilin family of actin-binding proteins. Structurally, twinfilins are characterized by two ADF/cofilin-like domains that share homology with the actin-depolymerizing factor (ADF)/cofilin proteins. While specific structural data for Xenopus Twinfilin-2-A is limited in the literature, twinfilins generally form a 1:1 complex with actin monomers . The protein likely shares significant sequence homology with twinfilins from other species, as twinfilins are highly conserved across eukaryotes from yeast to humans . For comparison, Xenopus ADF/cofilins (XACs) consist of 168 amino acids and show 77% identity to chick cofilin and 66% identity to chick ADF .
Twinfilin-2-A in Xenopus laevis functions as an actin monomer-sequestering protein that regulates actin turnover during embryonic development. Based on studies of Twinfilin-1 in Xenopus, which is essential for convergent extension movements during gastrulation, Twinfilin-2-A likely plays a significant role in controlling actin dynamics during morphogenetic movements . Mechanistically, twinfilins prevent actin filament assembly by binding to actin monomers and inhibiting their nucleotide exchange reaction . In developmental contexts, this activity helps maintain the proper balance between actin polymerization and depolymerization required for cellular processes such as lamellipodial dynamics and polarity establishment . Unlike cofilin/ADF proteins, twinfilins do not appear to directly induce actin filament depolymerization, suggesting a distinct regulatory mechanism .
For effective expression and purification of recombinant Xenopus laevis Twinfilin-2-A, researchers typically employ bacterial expression systems using E. coli, though yeast, baculovirus, or mammalian cell expression systems can also be utilized . A common methodological approach involves creating a GST-fusion construct by cloning the twf2-a coding sequence into a suitable expression vector.
For purification, the following protocol can be adapted from established methods for twinfilin proteins:
Express the GST-fusion protein in E. coli under optimal induction conditions.
Lyse cells and isolate the protein using glutathione-agarose beads.
Cleave the fusion protein using thrombin (typically 5 U/ml) through overnight incubation at 4°C.
Wash the beads with buffer containing 50 mM Tris (pH 7.5) and 150 mM NaCl.
Concentrate the supernatant using centrifugal filter devices with appropriate molecular weight cutoffs.
Further purify the protein by gel filtration chromatography using a Superdex-75 column equilibrated with 10 mM Tris (pH 7.5), 50 mM NaCl.
Pool peak fractions, concentrate to 30-100 μM, flash-freeze in liquid nitrogen, and store at -80°C .
The purified protein should demonstrate >85% purity as determined by SDS-PAGE analysis .
Recombinant Xenopus laevis Twinfilin-2-A serves as a valuable tool for investigating actin dynamics through several in vitro methodologies:
Actin Polymerization Assays: The effect of Twinfilin-2-A on actin filament assembly can be quantified by measuring the rate of pyrene-labeled actin polymerization in the presence of varying concentrations of the recombinant protein.
Co-sedimentation Assays: To assess binding affinity to actin monomers or filaments, mix purified Twinfilin-2-A with F-actin or G-actin at different ratios, followed by ultracentrifugation. Analyze the distribution of proteins between supernatant and pellet fractions using SDS-PAGE and densitometry .
Nucleotide Exchange Inhibition: Measure the inhibitory effect of Twinfilin-2-A on the exchange of bound nucleotide on actin monomers using fluorescently labeled nucleotides.
Single Filament Assays: Using total internal reflection fluorescence (TIRF) microscopy, visualize the effect of Twinfilin-2-A on individual actin filaments in real-time, providing insights into its role in filament dynamics.
These approaches allow for precise quantification of Twinfilin-2-A's biochemical activities and its concentration-dependent effects on actin dynamics.
Several methodological approaches are particularly effective for investigating Twinfilin-2-A function during Xenopus embryonic development:
Microinjection of mRNA or Morpholinos: Inject either synthetic twf2-a mRNA for overexpression or antisense morpholino oligonucleotides for knockdown into early Xenopus embryos (typically at the 1-4 cell stage). This allows for gain- or loss-of-function analysis in vivo.
Explant Culture Systems: Use Keller explants of the dorsal marginal zone (DMZ) to study convergent extension movements in controlled conditions. These explants can be manually dissected and cultured until desired developmental stages for analysis .
Fluorescent Speckle Microscopy: This technique involves microinjection of fluorescently labeled actin monomers at low concentrations to create speckles in the actin cytoskeleton. By tracking speckle lifetime, researchers can quantify actin turnover rates in the presence or absence of Twinfilin-2-A function .
Immunofluorescence Localization: Use specific antibodies against Twinfilin-2-A to visualize its distribution in embryonic tissues. This technique has revealed that related proteins like XAC show specific localization patterns during development, such as enrichment along membranes in the vegetal hemisphere after fertilization .
Time-lapse Imaging: Combine microinjection of fluorescently tagged Twinfilin-2-A with confocal microscopy to track protein dynamics during morphogenetic movements in real-time.
The methodological approaches for investigating Twinfilin-2-A function differ significantly between whole embryo and explant studies:
Whole Embryo Studies:
Allow assessment of global developmental phenotypes following Twinfilin-2-A manipulation.
Require careful staging according to Nieuwkoop and Faber (NF) developmental stages.
Often involve microinjection followed by whole-mount immunohistochemistry or in situ hybridization.
Phenotypic analysis typically focuses on major morphogenetic events like gastrulation and neurulation.
May use targeted injections (e.g., dorsal vs. ventral blastomeres) to assess tissue-specific functions.
Tissue Explant Studies:
Utilize manually dissected tissue fragments (e.g., dorsal marginal zone explants) cultured ex vivo.
Provide better optical access for high-resolution imaging of cellular behaviors.
Allow for more precise manipulation of experimental conditions (e.g., drug treatments).
Enable quantitative analysis of specific cell behaviors like protrusive activity or cell intercalation.
Often involve approximately 750 Keller explants cultivated until specific developmental stages (e.g., NF stage 14) .
The explant approach has been particularly valuable for studying actin regulatory proteins, as demonstrated by affinity purification mass spectrometry experiments that identified interaction partners of the related protein Cofilin2 specifically in cells undergoing convergent extension .
A comparative analysis reveals distinct functional characteristics among various actin-binding proteins in Xenopus laevis:
Twinfilin-2-A likely shares the actin monomer-sequestering activity with Twinfilin-1 but may have distinct expression patterns or developmental roles. Unlike Cofilin/XAC, twinfilins do not appear to directly induce actin filament depolymerization, despite sequence homology in their ADF/cofilin-like domains .
To effectively distinguish between the activities of Twinfilin-2-A and Twinfilin-2-B in Xenopus laevis, researchers can employ several targeted methodological approaches:
Isoform-Specific Antibodies: Develop and validate antibodies that specifically recognize unique epitopes in either Twinfilin-2-A or Twinfilin-2-B. These can be used for Western blotting, immunoprecipitation, and immunohistochemistry to determine expression patterns and subcellular localization differences .
RT-qPCR with Isoform-Specific Primers: Design primers that target unique regions in the mRNA sequences of twf2-a and twf2-b to quantify their relative expression levels across developmental stages and tissues.
Isoform-Selective Knockdown: Utilize morpholino oligonucleotides or CRISPR/Cas9 targeting strategies that selectively deplete either Twinfilin-2-A or Twinfilin-2-B, followed by phenotypic analysis to determine their specific functions.
Domain Swap Experiments: Create chimeric constructs that exchange domains between Twinfilin-2-A and Twinfilin-2-B to identify regions responsible for functional differences.
In Vitro Activity Assays: Compare the biochemical activities of purified recombinant Twinfilin-2-A and Twinfilin-2-B in various actin-related assays, including monomer sequestration, nucleotide exchange inhibition, and effects on polymerization/depolymerization rates.
These approaches can reveal functional specialization between these closely related isoforms during Xenopus development.
Twinfilin-2-A serves as a valuable molecular tool for investigating the actin-dependent mechanisms driving convergent extension (CE) during Xenopus gastrulation. Based on insights from studies of related proteins, several methodological approaches can be employed:
Loss-of-Function Analysis: Deplete Twinfilin-2-A using morpholino oligonucleotides or CRISPR/Cas9, followed by quantitative analysis of CE movements using Keller explants. This approach revealed that Twinfilin-1 loss disrupts lamellipodial dynamics and polarity, preventing proper convergent extension .
Rescue Experiments: After Twinfilin-2-A depletion, perform rescue experiments by co-injecting wild-type or mutant versions of the protein to identify functionally critical domains and residues.
Live Imaging of Actin Dynamics: Combine Twinfilin-2-A manipulation with fluorescent actin speckle microscopy to measure changes in actin turnover rates during CE. This methodology demonstrated that Twinfilin-1 is required for rapid actin turnover in the dorsal mesoderm, with its loss significantly increasing actin speckle lifetimes .
Analysis of Polarized Protrusions: Examine how Twinfilin-2-A affects the formation and dynamics of polarized protrusions during cell intercalation, which are critical for proper CE movements.
Investigation of Cytoplasmic Actin Cables: Determine whether Twinfilin-2-A, like Twinfilin-1, is involved in the assembly of polarized cytoplasmic actin cables that are essential for convergent extension .
These approaches can establish the specific contribution of Twinfilin-2-A to the complex cytoskeletal rearrangements driving morphogenetic movements during Xenopus development.
When faced with contradictory data regarding Twinfilin-2-A's role in actin dynamics, the following experimental approaches can provide clarity:
Concentration-Dependent Assays: Systematically test Twinfilin-2-A's activities across a wide concentration range in vitro, as some actin-binding proteins exhibit different activities at different concentrations. Document dose-response relationships for each activity.
Biochemical State Analysis: Investigate whether post-translational modifications (e.g., phosphorylation) alter Twinfilin-2-A's activity. This is particularly relevant given that Xenopus ADF/cofilin (XAC) exists in phosphorylated and dephosphorylated states with different activities during early development .
Context-Dependent Function Analysis: Examine whether Twinfilin-2-A's activity depends on the presence of other actin-binding proteins or specific cellular contexts. Co-immunoprecipitation followed by mass spectrometry can identify context-specific interaction partners.
Single-Molecule Approaches: Utilize single-molecule techniques to directly visualize individual Twinfilin-2-A molecules interacting with actin filaments or monomers in real-time, providing mechanistic insights that may resolve contradictions.
Cross-Validation with Multiple Techniques: Apply multiple independent methodologies to assess the same activity, such as combining bulk biochemical assays with microscopy-based approaches and cellular phenotypic analyses.
Comparative Analysis Across Species: Compare Twinfilin-2-A's activities across multiple model organisms to determine if contradictions reflect species-specific differences in function.
Investigation of Twinfilin-2-A phosphoregulation during development requires careful methodological considerations:
Phosphorylation Site Identification: Use mass spectrometry to map potential phosphorylation sites on Twinfilin-2-A. This approach can be informed by the analysis of related proteins like XAC, which is regulated through phosphorylation during early development .
Phosphorylation State-Specific Antibodies: Develop antibodies that specifically recognize phosphorylated or non-phosphorylated forms of Twinfilin-2-A for tracking its regulatory status during development.
Phosphomimetic and Phospho-Resistant Mutants: Create mutants with amino acid substitutions that either mimic (e.g., Ser/Thr to Asp/Glu) or prevent (e.g., Ser/Thr to Ala) phosphorylation at key sites. Test these in rescue experiments following endogenous protein depletion.
Temporal Analysis of Phosphorylation: Employ 2D Western blotting to track changes in phosphorylation states across developmental stages, similar to the approach that revealed rapid dephosphorylation of XAC after fertilization .
Kinase and Phosphatase Identification: Use inhibitor studies, co-immunoprecipitation, and in vitro kinase/phosphatase assays to identify enzymes regulating Twinfilin-2-A phosphorylation.
Functional Consequences: Assess how phosphorylation affects Twinfilin-2-A's interaction with actin monomers, actin filaments, and other binding partners through in vitro assays with phosphorylated and non-phosphorylated forms of the protein.
This approach is particularly relevant given that XAC exists exclusively in phosphorylated form in oocytes, with over 60% becoming dephosphorylated within 30 minutes after fertilization—a regulatory mechanism critical for proper cleavage during early development .
Producing functionally active recombinant Xenopus laevis Twinfilin-2-A presents several technical challenges:
| Challenge | Solution |
|---|---|
| Protein Solubility | Use solubility-enhancing fusion tags (GST, MBP); optimize expression temperature (typically 16-18°C); include solubility enhancers like sorbitol or arginine in buffers |
| Maintaining Native Conformation | Avoid harsh elution conditions; use gentle tag removal methods; include reducing agents to maintain proper disulfide bonding |
| Preventing Proteolytic Degradation | Add protease inhibitors throughout purification; minimize time between purification steps; maintain samples at 4°C |
| Assessing Functional Activity | Develop activity assays specific to Twinfilin-2-A; compare activity to positive controls (e.g., other twinfilins with known activity) |
| Protein Aggregation | Include low concentrations of non-ionic detergents in storage buffer; optimize salt concentration through stability screens |
| Long-term Storage Stability | Flash freeze in liquid nitrogen with 10-15% glycerol; store at -80°C; avoid repeated freeze-thaw cycles |
For optimal results, fusion proteins should be cleaved using site-specific proteases and further purified using gel filtration chromatography to achieve >85% purity as determined by SDS-PAGE analysis .
The pseudotetraploid nature of Xenopus laevis presents unique challenges for studying specific gene functions like Twinfilin-2-A. Researchers can address these challenges through several methodological approaches:
Homeolog-Specific Targeting: Design morpholinos, antisense oligonucleotides, or CRISPR/Cas9 guide RNAs that selectively target either the "a" or "b" homeolog of Twinfilin-2 by focusing on regions of sequence divergence.
Sequence Alignment Analysis: Perform careful sequence alignment of the homeologs to identify unique regions that can be used for selective manipulation or detection. This approach is informed by studies of XACs, which identified 12 amino acid differences between allelic variants spread throughout the sequence .
Homeolog Expression Profiling: Use RT-qPCR with homeolog-specific primers to determine the relative expression levels of twf2-a versus twf2-b across developmental stages and tissues, establishing which homeolog may be functionally dominant in specific contexts.
Complementation Studies: After depleting both homeologs, perform rescue experiments with each homeolog individually to assess functional redundancy or specialization.
Comparative Studies with Xenopus tropicalis: Utilize the diploid species Xenopus tropicalis, which has a single ortholog, as a complementary model for functional studies.
Considering Allelic Variation: Be aware that apparent homeologs might actually represent allelic variants of the same gene, as suggested for XACs based on similarities in developmental expression, embryonic localization, and non-coding sequence regions .
These strategies help disambiguate the functions of duplicate genes resulting from the ancestral whole-genome duplication event in X. laevis.
Several cutting-edge technologies hold promise for elucidating Twinfilin-2-A's role in embryonic development:
Optogenetic Control of Twinfilin-2-A Activity: Develop light-inducible systems to spatiotemporally control Twinfilin-2-A activity in developing embryos, enabling precise manipulation of actin dynamics in specific tissues or cells during morphogenesis.
Live Super-Resolution Microscopy: Apply techniques like Structured Illumination Microscopy (SIM) or Stochastic Optical Reconstruction Microscopy (STORM) to visualize Twinfilin-2-A interactions with the actin cytoskeleton at nanoscale resolution in living embryos.
Cryo-Electron Microscopy: Determine high-resolution structures of Twinfilin-2-A in complex with actin monomers or other binding partners to elucidate the molecular basis of its activities.
Single-Cell Transcriptomics and Proteomics: Profile gene expression and protein abundance at single-cell resolution to map Twinfilin-2-A expression patterns and identify co-regulated genes across developmental stages.
Genome-Wide CRISPR Screens: Identify genetic interactors of Twinfilin-2-A through systematic CRISPR/Cas9-mediated gene disruption followed by phenotypic analysis of morphogenetic movements.
Biomechanical Measurement Techniques: Combine Twinfilin-2-A manipulation with techniques like atomic force microscopy or optical tweezers to quantify how it influences cellular mechanical properties during morphogenesis.
These approaches would complement established methodologies like fluorescent speckle microscopy, which has already provided valuable insights into how related proteins like Twinfilin-1 control actin turnover during convergent extension .
Comparative studies of Twinfilin-2-A across species can inform therapeutic strategies targeting cytoskeletal dynamics in several ways:
Conservation Analysis for Drug Target Identification: Identifying highly conserved functional domains and residues in Twinfilin-2-A across species helps pinpoint potential therapeutic targets where intervention would likely have predictable effects across species, including humans.
Species-Specific Differences for Selective Targeting: Understanding species-specific differences in Twinfilin-2-A structure and function can guide the development of therapeutics that selectively target human proteins while minimizing off-target effects.
Evolutionary Insights into Functional Redundancy: Comparing the roles of Twinfilin-2-A across species with different levels of genetic redundancy (e.g., pseudotetraploid X. laevis versus diploid species) provides insights into potential compensatory mechanisms that might affect therapeutic efficacy.
Developmental Phenotypes as Predictors of Drug Effects: The developmental consequences of Twinfilin-2-A disruption in model organisms can predict potential adverse effects of therapeutics targeting this pathway in humans, particularly during embryonic development.
Cross-Species Validation of Mechanism: Confirming consistent mechanistic action of Twinfilin-2-A across diverse species strengthens the foundation for therapeutic development by suggesting evolutionary conservation of fundamental cytoskeletal regulatory pathways.