Recombinant Chicken Charged multivesicular body protein 2a (CHMP2A)

Shipped with Ice Packs
In Stock

Description

Introduction to Recombinant Chicken Charged Multivesicular Body Protein 2a (CHMP2A)

Recombinant Chicken Charged Multivesicular Body Protein 2a (CHMP2A) is a protein involved in various cellular processes, particularly in membrane remodeling and vesicle formation. While specific data on the recombinant chicken version might be limited, understanding its role in cellular biology can provide insights into its potential applications and functions.

CHMP2A is part of the ESCRT-III complex, which plays a crucial role in the endosomal sorting complex required for transport (ESCRT) pathway. This pathway is essential for processes such as multivesicular body (MVB) formation, cytokinesis, and autophagy. The ESCRT-III complex, including CHMP2A, is known for its ability to remodel membranes and facilitate membrane fission.

Membrane Remodeling and ESCRT-III Complex

CHMP2A, along with other ESCRT-III components, forms helical filaments that interact with membranes. These interactions are crucial for the constriction and cleavage of membranes, which are necessary for processes like MVB formation and autophagosome closure . The ESCRT-III complex, including CHMP2A, works in conjunction with the VPS4 ATPase to disassemble and recycle ESCRT-III components after membrane fission .

Autophagy

CHMP2A has been shown to play a significant role in autophagy, particularly in the closure of phagophores to form mature autophagosomes. Depletion of CHMP2A leads to the accumulation of unclosed phagophores, indicating impaired autophagic flux . This highlights the importance of CHMP2A in maintaining proper autophagic processes.

Potential Applications and Research Directions

Understanding the role of CHMP2A in membrane remodeling and autophagy can have implications for various biomedical applications:

  • Biotechnology: Recombinant proteins like CHMP2A could be used in biotechnological applications to study membrane dynamics or develop novel therapeutic strategies.

  • Disease Models: Given its role in autophagy and membrane remodeling, CHMP2A could serve as a model for studying diseases related to impaired autophagic processes or membrane dysfunction.

Data Tables and Research Findings

ProcessRole of CHMP2AKey Findings
Membrane RemodelingForms helical filaments with other ESCRT-III components to facilitate membrane constriction and cleavage.Essential for MVB formation and autophagosome closure .
AutophagyRequired for phagophore closure and autophagosome maturation.Depletion leads to accumulation of unclosed phagophores and impaired autophagic flux .
Structural InsightsAssembles into helical filaments with a positively charged surface for membrane interaction.Facilitates electrostatic interactions between filaments .

Product Specs

Form
Lyophilized powder
Lead Time
Delivery times vary depending on the purchasing method and location. Please consult your local distributor for specific delivery timelines. Note: All proteins are shipped with standard blue ice packs. Dry ice shipping requires advance notice and incurs additional charges.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to collect the contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting at -20°C/-80°C. Our standard glycerol concentration is 50%, which can serve as a reference.
Shelf Life
Shelf life depends on several factors, including storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized formulations have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt. Aliquot for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
The tag type is determined during manufacturing. If you require a specific tag, please inform us, and we will prioritize its development.
Synonyms
CHMP2A; RCJMB04_35d23Charged multivesicular body protein 2a; Chromatin-modifying protein 2a; CHMP2a
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-220
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Gallus gallus (Chicken)
Target Names
Target Protein Sequence
MDLLFGRRKT PEELLRQNQR ALTRAMRELD RERQKLEAQE KKIIIDIKKM AKQGQMDAVK IMAKDLVRTR RYVKKFITMR ANVQAVSLKI QTLKSNNSMA LAMKGVTKAM ATMNRQLKLP QIQKIMMEFE KQAGIMDMKE ELMNDAIDDA MGDEDDEEES DAVVSQVLDE LGLNLTDELA TLPPPGGSLA AGEGRAAEAA AALADADADL EERLKNLRRD
Uniprot No.

Target Background

Function
CHMP2A is a probable core component of the endosomal sorting complex required for transport III (ESCRT-III). ESCRT-III is involved in multivesicular body (MVB) formation and the sorting of endosomal cargo proteins into MVBs. MVBs contain intraluminal vesicles (ILVs) formed by invagination and scission from the endosomal limiting membrane. These ILVs are largely delivered to lysosomes for degradation of membrane proteins, such as stimulated growth factor receptors, lysosomal enzymes, and lipids.
Database Links
Protein Families
SNF7 family
Subcellular Location
Late endosome membrane; Peripheral membrane protein; Cytoplasmic side. Cytoplasm.

Q&A

What is chicken CHMP2A and what are its primary functions in cellular processes?

Chicken CHMP2A (Charged Multivesicular Body Protein 2a) is a member of the ESCRT-III complex that plays essential roles in membrane deformation and scission events in cells. Similar to mammalian systems, chicken CHMP2A likely functions in multivesicular body biogenesis, membrane abscission during cytokinesis, and membrane repair mechanisms. The ESCRT-III system recruits proteins through MIT:MIM (Microtubule Interacting and Transport:MIT-Interacting Motif) interactions, creating specialized assemblies that coordinate various cellular processes .

Why is recombinant chicken CHMP2A useful for research applications?

Recombinant chicken CHMP2A provides several advantages for research:

  • Avian expression systems can produce proteins with post-translational modifications that are more similar to human proteins compared to bacterial or yeast systems .

  • Studying CHMP2A in chickens provides evolutionary insights into ESCRT-III complex conservation and divergence.

  • Chickens serve as a valuable model organism for certain developmental and immunological studies where ESCRT-III function may be relevant.

  • Recombinant expression allows for protein engineering, including addition of tags for purification or visualization, and introduction of mutations to study structure-function relationships.

What are the key biological pathways involving chicken CHMP2A?

Chicken CHMP2A is involved in several critical cellular pathways:

  • Endosomal sorting and multivesicular body formation: CHMP2A participates in the sorting of ubiquitinated cargo proteins into intraluminal vesicles.

  • Cell division: ESCRT-III proteins, including CHMP2A, are recruited to the midbody during cytokinesis to facilitate membrane abscission .

  • Membrane repair: ESCRT-III components help seal damaged plasma membranes.

  • Viral budding: Many enveloped viruses hijack the ESCRT-III machinery, including CHMP2A, to facilitate viral release.

  • Autophagy: ESCRT-III proteins contribute to autophagosome formation and maturation.

How can we optimize the expression of recombinant chicken CHMP2A for structural studies?

Optimizing recombinant chicken CHMP2A expression for structural studies requires several strategic approaches:

  • Expression system selection: While bacterial systems like E. coli offer high yields, they lack appropriate post-translational modifications. For complete structural fidelity, consider using avian cell lines or genetically modified chickens that can produce proteins with native-like modifications .

  • Construct design:

    • Include a solubility tag (MBP, SUMO, or GST) at the N-terminus

    • Consider expressing truncated versions that remove the autoinhibitory C-terminal region to prevent aggregation

    • Include a cleavable His-tag for purification

    • Codon-optimize the sequence for the expression system

  • Purification strategy: Implement a multi-step purification protocol:

    • Initial capture using affinity chromatography (Ni-NTA for His-tagged proteins)

    • Ion exchange chromatography to separate charged variants

    • Size exclusion chromatography for final polishing and buffer exchange

  • Protein stability optimization:

    • Screen various buffers with different pH values (typically pH 7.2-8.0)

    • Test various salt concentrations (often 150-300 mM NaCl)

    • Include reducing agents like TCEP (0.5-1 mM) to maintain disulfide bonds

    • Consider adding glycerol (5-10%) to enhance stability

What are the challenges in studying CHMP2A interactions within the ESCRT-III complex in avian systems?

Studying CHMP2A interactions within the ESCRT-III complex in avian systems presents several challenges:

  • Transient nature of interactions: ESCRT-III components transition between closed monomeric and open polymeric states during assembly. These dynamic interactions can be difficult to capture experimentally.

  • Heterogeneity of complexes: ESCRT-III forms heteromeric filaments with variable stoichiometry depending on the specific cellular process, creating complexity in isolating discrete complexes.

  • Technical limitations:

    • Limited availability of chicken-specific antibodies for co-immunoprecipitation studies

    • Challenges in establishing stable chicken cell lines for protein-protein interaction studies

    • Difficulty in crystallizing complete ESCRT-III assemblies due to their polymeric nature

  • Functional redundancy: Potential redundancy among CHMP family proteins may mask phenotypes in knockout or knockdown studies.

  • Context-dependent assembly: ESCRT-III assembly is influenced by membrane composition, curvature, and specific recruiting factors, making in vitro reconstitution challenging.

To address these challenges, researchers often combine multiple approaches including proteomics analysis, gene editing technologies like CRISPR-Cas9, and advanced imaging techniques such as super-resolution microscopy or cryo-EM .

How does post-translational modification affect chicken CHMP2A function and how can these modifications be preserved in recombinant systems?

Post-translational modifications (PTMs) significantly impact CHMP2A function by regulating its:

  • Conformational switching: Phosphorylation can destabilize the autoinhibited conformation, promoting ESCRT-III assembly.

  • Protein-protein interactions: PTMs can create or disrupt binding sites for interaction partners.

  • Subcellular localization: Certain modifications direct CHMP2A to specific cellular compartments.

  • Dissociation from membranes: ATP-dependent processes involving VPS4 require proper modifications of ESCRT-III components.

To preserve these modifications in recombinant systems:

  • Select appropriate expression platforms:

    • Transgenic chicken systems or avian cell lines maintain most relevant PTMs

    • Mammalian cell lines (HEK293T, HeLa) can provide many native modifications

    • Avoid bacterial expression if PTMs are critical

  • Targeted modification strategies:

    • Co-express relevant kinases or other modifying enzymes

    • Use chemical biology approaches for site-specific modifications

    • Consider enzymatic in vitro modification after purification

  • Verification methods:

    • Mass spectrometry analysis to confirm modification patterns

    • Functional assays comparing recombinant proteins to native counterparts

    • Structural analyses to verify proper folding

The chicken as an expression system has advantages for human protein production as chickens do not produce α1,3-Gal epitopes (unlike mammalian systems), which can cause immunological reactions. This makes chickens potentially valuable for producing recombinant proteins with more human-compatible glycosylation patterns .

What gene editing strategies are most effective for studying chicken CHMP2A in vivo?

Several gene editing strategies have proven effective for studying chicken CHMP2A function in vivo:

  • CRISPR-Cas9 knockin approaches:

    • Precise editing of the CHMP2A locus to introduce tags or reporter genes

    • Generation of conditional knockout models using inducible systems

    • Introduction of specific mutations to study structure-function relationships

  • Transgenic reporter systems:

    • Similar to the XCR1-iCaspase9-RFP system described in the literature, where the endogenous gene is replaced with a construct containing fluorescent proteins and inducible elements

    • This allows visualization of expression patterns and conditional ablation studies

  • Viral vector delivery systems:

    • Lentiviral or retroviral vectors for stable integration of modified CHMP2A constructs

    • Useful for both cultured cells and in vivo applications through embryo manipulation

  • Primordial germ cell (PGC) manipulation:

    • Chicken-specific approach where PGCs are isolated, genetically modified, and reintroduced to develop transgenic chickens

    • Particularly useful for studying developmental roles of CHMP2A

These approaches can be combined with modern imaging techniques and proteomics analysis to comprehensively characterize CHMP2A function in chicken models .

What purification strategies yield the highest quality recombinant chicken CHMP2A protein?

Obtaining high-quality recombinant chicken CHMP2A requires a strategic purification approach:

Step 1: Optimized expression system

  • Mammalian cells (HEK293T) provide good yield with proper folding and modifications

  • Insect cells (Sf9/Hi5) balance yield and post-translational modifications

  • Avian cell lines maintain species-specific modifications

Step 2: Multi-step purification protocol

  • Initial capture: Affinity chromatography using:

    • Streptactin resin for StrepII-tagged constructs (eluted with desthiobiotin)

    • Ni-NTA for His-tagged constructs (eluted with imidazole gradient)

    • Anti-FLAG for FLAG-tagged constructs

  • Intermediate purification:

    • Ion exchange chromatography (MonoQ or MonoS depending on isoelectric point)

    • Removal of contaminating nucleic acids and similarly charged proteins

  • Polishing step:

    • Size exclusion chromatography using Superdex 200 column in buffer containing:

      • 20 mM Tris (pH 7.2 at 23°C)

      • 150 mM NaCl

      • 0.5 mM TCEP

Step 3: Quality assessment

  • SDS-PAGE and western blotting to confirm purity and identity

  • Mass spectrometry to verify sequence and modifications

  • Dynamic light scattering to assess homogeneity

  • Functional assays to confirm activity

Critical considerations

  • CHMP2A tends to aggregate when the autoinhibitory C-terminal domain is disrupted

  • Addition of stabilizers (5-10% glycerol, 1 mM DTT) helps maintain protein stability

  • Flash freezing in small aliquots prevents degradation during storage

How can researchers effectively design co-immunoprecipitation experiments to study chicken CHMP2A interactions?

Designing effective co-immunoprecipitation (co-IP) experiments for chicken CHMP2A requires careful consideration of several factors:

Experimental design considerations:

  • Epitope tag selection:

    • N-terminal tags are preferred since C-terminal tags may interfere with MIM elements

    • Common options include FLAG, Myc, or StrepII tags

    • Consider dual tagging strategies for tandem affinity purification

  • Cell lysis conditions:

    • Use mild detergents (0.5% TritonX-100) to preserve protein-protein interactions

    • Include protease inhibitor cocktails to prevent degradation

    • Maintain physiological salt concentrations (150 mM NaCl)

    • Consider crosslinking (1% formaldehyde) for transient interactions

  • Antibody selection and validation:

    • Test antibody specificity in chicken tissues/cells

    • Consider using anti-tag antibodies if chicken-specific antibodies are unavailable

    • Validate with western blotting before co-IP

  • Controls to include:

    • Input sample (pre-immunoprecipitation lysate)

    • IgG control (non-specific antibody)

    • Lysate from cells not expressing the tagged protein

    • Competitive binding with peptides containing MIM motifs

Protocol optimization tips:

  • Adjust salt and detergent concentrations to balance specificity and sensitivity

  • Test various incubation times (1-4 hours) and temperatures (4°C is standard)

  • For ESCRT-III components, consider membrane fractionation to enrich for active complexes

  • Use nuclease treatment to eliminate DNA/RNA-mediated interactions

Analysis methods:

  • Standard western blotting for known interactors

  • Mass spectrometry (LC-MS/MS) for unbiased identification of novel binding partners

  • Quantitative proteomics using SILAC or TMT labeling for comparative interaction studies

Example co-IP protocol:
Similar to protocols used for ESCRT-III components where cells are harvested 48 hours post-transfection, lysed in cold buffer (50 mM Tris pH 7.2, 150 mM NaCl, 0.5% TritonX-100, 1 mM DTT with protease inhibitors), clarified by centrifugation, and incubated with affinity resin for 1 hour at 4°C .

What imaging techniques are most suitable for visualizing chicken CHMP2A dynamics in live cells?

Visualizing chicken CHMP2A dynamics in live cells requires advanced imaging techniques that balance spatial resolution, temporal sensitivity, and minimal phototoxicity:

Fluorescence-based techniques:

  • Confocal microscopy with fluorescent protein fusions:

    • Tag CHMP2A with monomeric fluorescent proteins (mEGFP, mCherry)

    • Benefits: Widely accessible, good for tracking general localization

    • Limitations: Diffraction-limited resolution (~250 nm), potential functional interference

    • Best for: Tracking CHMP2A recruitment to endosomes or the midbody

  • Super-resolution microscopy:

    • Techniques include:

      • Structured Illumination Microscopy (SIM): 2× resolution improvement with good temporal resolution

      • Stimulated Emission Depletion (STED): Higher resolution but more phototoxic

      • PALM/STORM: Highest resolution but slower acquisition

    • Benefits: Resolves ESCRT-III filament organization and substructure

    • Limitations: Higher technical complexity, potential photobleaching

    • Best for: Detailed structures of ESCRT-III assemblies at the midbody or endosomes

  • Lattice light-sheet microscopy:

    • Benefits: Reduced phototoxicity, excellent for long-term imaging

    • Limitations: Specialized equipment requirements

    • Best for: Tracking CHMP2A throughout cell division or endosome maturation

Complementary approaches:

  • CRISPR-Knock-in fluorescent tagging:

    • Similar to approaches used for XCR1 proteins in chickens, where endogenous loci are tagged with fluorescent proteins

    • Benefits: Physiological expression levels, improved functional preservation

    • Example: Generation of CHMP2A-TagRFP knock-in chickens using CRISPR-Cas9

  • Split fluorescent protein complementation:

    • Tag CHMP2A and interaction partners with complementary fragments

    • Benefits: Visualization of specific protein-protein interactions

    • Limitations: Irreversible complementation can affect dynamics

  • FRAP (Fluorescence Recovery After Photobleaching):

    • Benefits: Measures mobility and exchange rates of CHMP2A in complexes

    • Applications: Determine assembly/disassembly kinetics at different cellular locations

Important considerations:

  • Ensure fluorescent tags don't disrupt MIT:MIM interactions critical for ESCRT-III function

  • Validate tagged CHMP2A functionality through rescue experiments

  • Include appropriate controls (expression levels, photobleaching, fixation artifacts)

  • Combine multiple approaches for comprehensive characterization

How can quantitative proteomics be applied to study changes in CHMP2A-associated proteins under different conditions?

Quantitative proteomics offers powerful approaches to study CHMP2A-associated proteins and how these interactions change under different biological conditions:

Experimental approaches:

  • Immunoprecipitation-Mass Spectrometry (IP-MS):

    • Immunoprecipitate CHMP2A from cells under different conditions

    • Digest proteins and analyze peptides by LC-MS/MS

    • Quantify differences in protein abundances

    • Similar to approaches used for other protein complexes

  • Proximity labeling techniques:

    • BioID: Fuse CHMP2A with a promiscuous biotin ligase

    • APEX2: Fuse CHMP2A with an engineered peroxidase

    • TurboID: Faster labeling kinetics for capturing dynamic interactions

    • These enzymes tag neighboring proteins, which are then purified and identified by MS

  • Quantification strategies:

    • Label-free quantification: Compare peptide intensities across samples

    • SILAC (Stable Isotope Labeling with Amino acids in Cell culture): Metabolic labeling

    • TMT (Tandem Mass Tag) or iTRAQ: Chemical labeling for multiplexed analysis

    • Spectral counting: Simpler but less accurate quantification

Data analysis and interpretation:

  • Statistical analysis:

    • Apply appropriate statistical tests (t-tests, ANOVA)

    • Use fold-change thresholds (typically >2.0 or <0.5) with significance cutoffs (p≤0.05)

    • Implement multiple testing corrections (FDR)

  • Network analysis:

    • Generate protein-protein interaction networks

    • Identify condition-specific interaction modules

    • Perform Gene Ontology enrichment analysis

  • Validation strategies:

    • Co-immunoprecipitation and western blotting for selected interactors

    • Microscopy colocalization studies

    • Functional assays to test biological relevance

Application examples:

Research QuestionProteomics ApproachExpected Outcome
CHMP2A interactome during cytokinesisBioID tagging combined with cell synchronization and TMT labelingIdentification of cell cycle-specific ESCRT-III regulators
Effect of viral infection on CHMP2A complexesIP-MS of CHMP2A before and after viral infection with SILACDetection of viral proteins that hijack ESCRT machinery
Tissue-specific CHMP2A partnersIP-MS from different chicken tissues with label-free quantificationTissue-specific regulators of ESCRT-III function
PTM-dependent interactionsIP-MS with phosphatase treatment vs. untreated samplesPhosphorylation-dependent binding partners

This approach successfully identified 61 differentially expressed proteins in studies of recombinant influenza viruses, demonstrating the power of quantitative proteomics for identifying condition-specific protein changes .

What assays can be used to evaluate the membrane remodeling activity of recombinant chicken CHMP2A?

Several in vitro and cellular assays can be employed to assess the membrane remodeling capabilities of recombinant chicken CHMP2A:

In vitro membrane remodeling assays:

  • Giant Unilamellar Vesicle (GUV) deformation assays:

    • Purified recombinant CHMP2A is incubated with fluorescently labeled GUVs

    • Membrane deformation is visualized using confocal microscopy

    • Quantification: Measure tubulation frequency, tube dimensions, and vesiculation events

    • Controls: Inactive CHMP2A mutants, other ESCRT-III proteins

  • Supported lipid bilayer (SLB) assays:

    • CHMP2A polymerization on supported membranes

    • Visualization using atomic force microscopy or TIRF microscopy

    • Quantification: Filament dimensions, organization patterns, and membrane deformation

  • Liposome binding and tubulation assays:

    • Incubate recombinant CHMP2A with liposomes of defined composition

    • Assess binding by co-sedimentation or flotation assays

    • Visualize membrane tubulation by negative-stain electron microscopy

Cellular assays:

  • Dominant-negative overexpression:

    • Express mutant CHMP2A (e.g., MIT domain mutations) in chicken cell lines

    • Assess endosome morphology, cytokinesis completion, and viral budding

    • Quantification: Multinucleation, enlarged endosomes, reduced viral production

  • CRISPR-Cas9 knockout/knockin approaches:

    • Generate CHMP2A knockout cells/animals with conditional rescue

    • Similar to approaches used for XCR1 studies in chickens

    • Assess cellular phenotypes across different tissues/conditions

  • Live cell imaging with membrane probes:

    • Co-express fluorescently tagged CHMP2A with membrane markers

    • Monitor membrane dynamics during endosome maturation or cell division

    • Quantification: Membrane scission timing, efficiency, and morphology

Reconstitution systems:

  • Minimal ESCRT-III assembly systems:

    • Combine recombinant CHMP proteins in defined stoichiometry

    • Assess polymerization and membrane remodeling activity

    • Test contributions of different domains through truncation or mutation

  • Cell-free reactions:

    • Use cell extracts supplemented with recombinant proteins

    • Monitor specific processes (e.g., MVB formation, cytokinesis)

    • Deplete endogenous proteins and rescue with recombinant versions

These assays should be performed with appropriate controls, including inactive mutants and related ESCRT-III proteins, to establish specificity and functional significance of the observed activities.

How can researchers determine if chicken CHMP2A functionally complements human CHMP2A in cellular processes?

Determining functional complementation between chicken and human CHMP2A requires systematic approaches that assess the ability of chicken CHMP2A to rescue defects caused by human CHMP2A depletion:

Knockout/knockdown-rescue experiments:

  • CRISPR-Cas9 knockout or RNAi knockdown of human CHMP2A:

    • Generate human cell lines lacking endogenous CHMP2A

    • Alternatively, use siRNA/shRNA targeting untranslated regions of human CHMP2A

    • Document the resulting phenotypes (cytokinesis failure, defective MVB formation)

  • Rescue with chicken CHMP2A:

    • Express chicken CHMP2A in human CHMP2A-depleted cells

    • Use expression vectors with different promoters to test dose-dependency

    • Include appropriate controls (human CHMP2A rescue, inactive mutants)

  • Quantitative phenotypic analysis:

    • Measure restoration of normal cytokinesis (reduction in multinucleated cells)

    • Assess MVB formation and cargo sorting using electron microscopy

    • Evaluate viral budding efficiency for relevant viruses

    • Compare rescue efficiency of chicken vs. human CHMP2A

Domain swap experiments:

  • Generate chimeric CHMP2A proteins:

    • Create fusion proteins combining domains from chicken and human CHMP2A

    • Express in human CHMP2A-depleted cells

    • Identify which domains are interchangeable vs. species-specific

  • Focus on critical interaction interfaces:

    • MIT domains that mediate interactions with VPS4

    • MIM elements involved in ESCRT-III assembly

    • Membrane-binding regions

Biochemical interaction analysis:

  • Co-immunoprecipitation assays:

    • Test if chicken CHMP2A interacts with human ESCRT-III partners

    • Employ approaches similar to those described for ESCRT-III components

    • Compare interaction profiles of human and chicken CHMP2A

  • In vitro reconstitution:

    • Purify recombinant human and chicken CHMP2A

    • Assess polymer formation with other ESCRT-III components

    • Test ATPase stimulation of human VPS4

Quantitative comparison table:

Functional PropertyAssay MethodExpected Outcome for Full Complementation
Cytokinesis completionTime-lapse imagingReduction in multinucleated cells to wild-type levels
MVB biogenesisElectron microscopy, EGF degradation assaysNormal MVB morphology and degradation kinetics
Viral buddingViral titer measurementRestoration of efficient virus production
ESCRT-III assemblyFRET assays, super-resolution microscopyFormation of functional ESCRT-III polymers
VPS4 recruitmentCo-localization analysisProper recruitment to sites of membrane remodeling

This systematic approach will determine whether chicken CHMP2A can functionally replace human CHMP2A or if there are species-specific differences that affect certain ESCRT-III functions.

What are the critical controls needed when studying the effects of CHMP2A mutations on virus budding in avian cells?

When investigating the effects of CHMP2A mutations on virus budding in avian cells, several critical controls are essential to ensure data integrity and meaningful interpretation:

Genetic and expression controls:

  • Wild-type CHMP2A control:

    • Include wild-type chicken CHMP2A expression as positive control

    • Match expression levels between wild-type and mutant constructs

    • Use quantitative western blotting to verify expression levels

  • Expression level titration:

    • Test multiple expression levels to prevent artifacts from overexpression

    • Use inducible expression systems (such as doxycycline-inducible promoters)

    • Include dose-response analysis for phenotypic effects

  • Knockdown efficiency verification:

    • For RNAi experiments, quantify knockdown efficiency by qRT-PCR and western blot

    • Include rescue controls with RNAi-resistant constructs

    • For CRISPR-edited cells, verify genomic modifications by sequencing

Functional controls:

  • Known functional mutants:

    • Include previously characterized mutations with established phenotypes

    • Test CHMP2A with mutations in MIT domains or MIM elements

    • Use mutations that disrupt membrane binding as negative controls

  • Other ESCRT pathway controls:

    • Test effects of disrupting different ESCRT-III components

    • Include VPS4 dominant-negative constructs as a positive control for budding defects

    • Test ALIX depletion to distinguish ESCRT-pathway dependency

  • Non-ESCRT-dependent virus control:

    • Include viruses known to bud independently of ESCRT machinery

    • Compare with viruses having established ESCRT-dependency

    • This distinguishes specific vs. non-specific effects on viral replication

Virus-specific controls:

  • Multiple virus types:

    • Test effects on different enveloped viruses (influenza, retrovirus, etc.)

    • Compare with non-enveloped viruses as negative controls

    • This approach was effectively used in recombinant influenza virus studies

  • Viral component analysis:

    • Assess both viral RNA/DNA and protein production

    • Distinguish between assembly defects vs. budding defects

    • Measure intracellular vs. extracellular viral components

Imaging and ultrastructural controls:

  • Sub-cellular localization controls:

    • Verify proper localization of CHMP2A constructs

    • Include membrane and organelle markers

    • Compare wild-type and mutant localization patterns

  • Electron microscopy controls:

    • Examine ultrastructure of budding virions

    • Quantify arrested budding structures vs. completed virion release

    • Include both thin-section and immuno-EM approaches

Experimental design table:

Control TypeSpecific ControlPurpose
GeneticWild-type CHMP2AEstablish baseline function
GeneticEmpty vectorControl for transfection effects
GeneticDominant negative VPS4Positive control for ESCRT pathway disruption
ExpressionMatched expression levelsPrevent artifacts from variable expression
ExpressionInducible expressionTest dose-dependent effects
ViralESCRT-dependent virus (e.g., HIV)Test specific ESCRT function
ViralESCRT-independent virusSpecificity control
CellularCell viability assessmentRule out cytotoxicity as cause of effects
ImagingCo-localization with viral componentsVerify relevant interactions
BiochemicalInteraction verificationConfirm predicted molecular consequences of mutations

By incorporating these controls, researchers can confidently attribute observed phenotypes to specific molecular functions of CHMP2A and distinguish between direct and indirect effects on viral budding.

What are the major challenges in expressing and purifying functional recombinant chicken CHMP2A and how can they be overcome?

Expressing and purifying functional recombinant chicken CHMP2A presents several significant challenges, each requiring specific strategies to overcome:

Challenge 1: Protein aggregation and insolubility

ESCRT-III proteins like CHMP2A naturally polymerize, making them prone to aggregation when overexpressed:

Solutions:

  • Express fusion proteins with solubility-enhancing tags (MBP, SUMO)

  • Maintain the autoinhibited conformation by including the C-terminal region

  • Use low induction temperatures (16-18°C) for bacterial expression

  • Include mild detergents (0.05% DDM) during purification

  • Purify under denaturing conditions followed by controlled refolding

  • Consider co-expression with binding partners that stabilize monomeric forms

Challenge 2: Preserving native conformation and activity

CHMP2A function depends on its ability to transition between closed and open conformations:

Solutions:

  • Verify activity with in vitro polymerization assays

  • Employ membrane binding assays to confirm functionality

  • Use circular dichroism to monitor secondary structure integrity

  • Develop activity-based probes to assess conformational state

  • Include stabilizing buffers with physiological salt conditions (150 mM NaCl) and reducing agents (0.5 mM TCEP)

Challenge 3: Low expression yields in eukaryotic systems

Higher eukaryotic expression systems that preserve proper folding often produce lower yields:

Solutions:

  • Optimize codon usage for the expression system

  • Use strong promoters with inducible control

  • Implement high-cell-density cultivation strategies

  • Consider baculovirus expression systems for improved yields

  • For mammalian expression, use suspension-adapted HEK293 cells

  • In chicken systems, use egg white for secreted proteins

Challenge 4: Proper post-translational modifications

CHMP2A function may depend on specific modifications:

Solutions:

  • Select expression systems that provide relevant modifications

  • Characterize modifications using mass spectrometry

  • For phosphorylation-dependent functions, express in cells with active kinases

  • Consider chicken-derived expression systems for avian-specific modifications

Challenge 5: Purification homogeneity

CHMP2A may exist in multiple oligomeric states:

Solutions:

  • Implement multi-step purification strategies

  • Use size exclusion chromatography as final polishing step

  • Consider native gel electrophoresis to assess oligomeric state

  • Apply analytical ultracentrifugation to characterize protein complexes

  • Use single-molecule techniques to assess conformational heterogeneity

Comprehensive purification workflow:

StageMethodKey ParametersQuality Control
ExpressionMammalian cells (HEK293T)48h post-transfection, 37°C, 5% CO₂ Western blot verification
Initial lysisDetergent extraction50 mM Tris pH 7.2, 150 mM NaCl, 0.5% TritonX-100, protease inhibitors Bradford assay for yield
Affinity captureStreptactin resin for StrepII tag1h incubation at 4°C with gentle rotation SDS-PAGE analysis
Intermediate purificationIon exchangeSalt gradient elutionPurity assessment
Final polishingSize exclusionSuperdex 200 column, 0.5 mL/min flow rate Dynamic light scattering
StorageFlash freezingSmall aliquots, -80°CActivity retention testing

How can researchers troubleshoot inconsistent results when studying CHMP2A function in different experimental systems?

Inconsistent results when studying CHMP2A function across different experimental systems is a common challenge that requires systematic troubleshooting approaches:

Source of variation 1: Expression level differences

Inconsistent CHMP2A expression levels can dramatically impact phenotypic outcomes:

Troubleshooting strategies:

  • Implement quantitative western blotting to normalize expression levels

  • Use fluorescence-based protein quantification in live cells

  • Develop inducible expression systems with titratable control

  • Include internal standards for normalization across experiments

  • Perform dose-response experiments to understand threshold effects

Source of variation 2: Cell type-specific factors

CHMP2A function may depend on cell type-specific cofactors or regulators:

Troubleshooting strategies:

  • Compare results across multiple cell types systematically

  • Identify cell-specific interaction partners through proteomics

  • Consider genetic background differences that may influence results

  • Supplement deficient cells with factors from permissive cells

  • Use primary cells where possible to avoid cell line artifacts

Source of variation 3: Assay conditions and timing

ESCRT-III function is temporally regulated and condition-dependent:

Troubleshooting strategies:

  • Standardize experimental timelines and conditions

  • Implement time-course experiments to capture dynamic processes

  • Control for cell cycle stage through synchronization

  • Establish consistent environmental conditions (temperature, pH, media composition)

  • Include time-matched controls for all experimental conditions

Source of variation 4: Protein tagging and modification effects

Different tags or construct designs can affect CHMP2A function:

Troubleshooting strategies:

  • Compare multiple tagging strategies (N-terminal vs. C-terminal)

  • Test tag-free versions where possible

  • Use small epitope tags (FLAG, HA) that minimize functional interference

  • Validate tagged constructs with rescue experiments

  • Consider tag position effects on protein-protein interactions

Source of variation 5: Methodological differences

Different detection methods can yield apparently contradictory results:

Troubleshooting strategies:

  • Apply multiple orthogonal techniques to the same biological question

  • Standardize image acquisition and analysis parameters

  • Implement blinded analysis to prevent bias

  • Develop quantitative metrics for phenotype assessment

  • Include cross-laboratory validation when possible

Comprehensive troubleshooting workflow:

IssueDiagnostic ApproachPotential Solution
Inconsistent phenotypesTitrate expression levelsEstablish dose-response relationship
Cell type-specific effectsComparative proteomicsIdentify missing cofactors
Temporal variationTime-course experimentsDetermine optimal time points
Construct-dependent resultsCompare multiple constructsIdentify minimally disruptive design
Conflicting interaction dataUse multiple interaction assaysFocus on interactions detected by multiple methods
Species-specific differencesDomain-swap experimentsMap functionally divergent regions

Standardization approaches:

  • Develop shared resource centers for validated reagents

  • Establish detailed protocols with explicit quality control metrics

  • Implement reporting standards that include key experimental parameters

  • Create reference datasets for benchmarking new experimental approaches

  • Use quantitative statistical methods to assess reproducibility across experiments

By systematically addressing these sources of variation, researchers can distinguish between genuine biological insights and technical artifacts when studying CHMP2A function.

What are the most promising future directions for research on recombinant chicken CHMP2A?

The exploration of recombinant chicken CHMP2A offers several exciting research avenues that could significantly advance our understanding of ESCRT-III biology and enable novel biotechnological applications:

  • Comparative structural biology: Determining high-resolution structures of chicken CHMP2A in different conformational states would provide valuable insights into the evolutionary conservation of ESCRT-III mechanisms. Recent advances in cryo-EM make it feasible to visualize ESCRT-III assemblies at near-atomic resolution, potentially revealing species-specific adaptations in polymer architecture .

  • Gene-edited chicken models: Developing CRISPR-engineered chicken lines with tagged or modified CHMP2A would enable in vivo studies of ESCRT-III function in avian development and physiology. Similar approaches have been successfully implemented for other proteins in chickens, such as the XCR1-iCaspase9-RFP system .

  • Viral-host interactions: Investigating how avian viruses interact with chicken ESCRT-III components could reveal important species-specific mechanisms of viral budding and host restriction. This is particularly relevant for zoonotic viruses that cross between avian and mammalian hosts, such as influenza .

  • Therapeutic protein production: Exploring the potential of genetically modified chickens as bioreactors for producing therapeutic proteins that require specific post-translational modifications. The advantages of chicken-based expression systems, including their inability to produce immunogenic α1,3-Gal epitopes, make them attractive alternatives to mammalian systems for certain applications .

  • Evolutionary biology of membrane remodeling: Comparative studies between chicken and mammalian ESCRT systems could reveal how this essential cellular machinery has evolved across vertebrate lineages while maintaining core functions in membrane remodeling.

  • Development of species-specific research tools: Creating chicken-specific antibodies, biosensors, and other research tools would facilitate more detailed investigations into avian-specific aspects of ESCRT-III biology that may differ from mammalian systems.

  • Cross-species complementation studies: Systematic analysis of functional complementation between chicken and human ESCRT-III components could identify critical species-specific interaction interfaces and regulatory mechanisms.

These research directions would not only advance our fundamental understanding of ESCRT-III biology but could also lead to practical applications in biotechnology, virology, and biomedicine. The combination of structural biology, gene editing, and functional genomics approaches offers powerful ways to explore the unique features of chicken CHMP2A while placing them in an evolutionary context.

What are the key takeaways for researchers beginning work with recombinant chicken CHMP2A?

For researchers beginning work with recombinant chicken CHMP2A, several key considerations will help ensure successful experimental outcomes:

  • Expression system selection is critical: Choose expression systems based on your experimental needs. Bacterial systems provide high yields but lack post-translational modifications, while avian systems maintain native modifications but with lower yields. For structural studies, E. coli expression with solubility tags may be sufficient, but functional studies typically require eukaryotic expression systems .

  • Protein design matters: CHMP2A transitions between closed and open conformations, with the open form prone to polymerization. Include the C-terminal autoinhibitory region for stability during purification, and consider tag placement carefully to avoid interfering with functional domains. N-terminal tags are generally preferred over C-terminal tags that might disrupt MIM elements .

  • Quality control is essential: Verify protein folding and activity before conducting advanced experiments. Simple assays like membrane binding can confirm basic functionality. For more complex activities, in vitro reconstitution with other ESCRT-III components can validate assembly competence.

  • Comparative approach yields insights: When possible, conduct parallel experiments with mammalian CHMP2A to identify conserved and divergent features. This comparative approach can reveal fundamental versus species-specific aspects of ESCRT-III biology.

  • Context matters: ESCRT-III proteins function in various cellular processes with different co-factors. When studying a specific process (e.g., viral budding, cytokinesis), ensure the relevant components of the pathway are present and functioning.

  • Validation through multiple methods: Confirm key findings through orthogonal techniques. For interaction studies, complement co-immunoprecipitation with microscopy colocalization or functional rescue experiments .

  • Consider evolutionary context: Chicken CHMP2A represents an evolutionary intermediate between mammalian and more distant vertebrate models. This position makes it valuable for understanding both conserved core functions and adaptive specializations in the ESCRT system.

  • Start with established protocols: Build upon published methods for related proteins. Purification approaches used for human ESCRT-III components often translate well to chicken orthologs, with minor optimizations for species-specific properties .

  • Document everything: ESCRT-III proteins can be challenging to work with, so detailed documentation of conditions, including buffer compositions, expression conditions, and purification parameters, is essential for troubleshooting and reproducibility.

  • Collaborate across disciplines: The complexity of ESCRT biology benefits from collaborative approaches combining structural biology, cell biology, virology, and evolutionary biology perspectives. Establishing collaborations can accelerate progress and provide complementary expertise.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.