Chmp2a, or Charged Multivesicular Body Protein 2a, in Danio rerio (Zebrafish) is a protein that is a probable core component of the endosomal sorting required for transport complex III (ESCRT-III) . The human form of the protein is also known as chromatin-modifying protein 2A .
| Gene Symbol | CHMP2A |
|---|---|
| Entrez Gene ID | 27243 |
| Full Name | Charged multivesicular body protein 2A |
| Synonyms | BC-2, BC2, CHMP2, VPS2, VPS2A |
| Gene Type | protein-coding |
| Organism | Homo sapiens (human) |
Chmp2a is involved in the formation of multivesicular bodies (MVBs) and the sorting of endosomal cargo proteins into MVBs . MVBs contain intraluminal vesicles (ILVs) that are generated by invagination and scission from the limiting membrane of the endosome and are mostly delivered to lysosomes, enabling the degradation of membrane proteins, such as stimulated growth factor receptors, lysosomal enzymes, and lipids . The MVB pathway requires the sequential function of ESCRT-0, -I, -II, and -III complexes . ESCRT-III proteins mostly dissociate from the invaginating membrane before the ILV is released . The ESCRT machinery also functions in topologically equivalent membrane fission events, such as the terminal stages of cytokinesis . Together with SPAST, the ESCRT-III complex promotes nuclear envelope sealing and mitotic spindle disassembly during late anaphase .
Chmp2a is a key component of the ESCRT-III complex, which is essential for endosomal sorting . Endosomal sorting is crucial for the degradation of surface receptor proteins and the formation of endocytic multivesicular bodies (MVBs) . ESCRT-III complexes are involved in membrane budding, endosome organization, and endosomal transport . They also participate in multivesicular body organization and assembly . Furthermore, Chmp2a participates in ESCRT complex disassembly .
The CHMP2A gene has been identified as potentially relevant in endometrial carcinoma (EC) . Studies suggest that the methylation status of CHMP2A is correlated with its expression levels in EC samples .
Chmp2a interacts with several other proteins to perform its function. These include :
Chmp1a: Charged multivesicular body protein 1a
Chmp1b: Charged multivesicular body protein 1b
Chmp3: Charged multivesicular body protein 3
Vps4a: Vacuolar protein sorting 4 homolog A
Chmp4bb: Charged multivesicular body protein 4c
Ist1: IST1 factor-associated with ESCRT-III
Chmp5b: Charged multivesicular body protein 5
Chmp7: Charged multivesicular body protein 7
Chmp6b: Charged multivesicular body protein 6
Chmp4c: Charged multivesicular body protein 4c
Danio rerio Charged multivesicular body protein 2a (chmp2a) is a likely 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) generated by invagination and scission from the endosomal limiting membrane. These ILVs are primarily delivered to lysosomes, facilitating the degradation of membrane proteins, such as stimulated growth factor receptors, lysosomal enzymes, and lipids.
CHMP2A (Charged Multivesicular Body Protein 2A) is a protein coding gene belonging to the chromatin-modifying protein/charged multivesicular body protein family. In zebrafish, as in other vertebrates, CHMP2A is a critical component of the ESCRT-III (endosomal sorting complex required for transport III) complex. This complex is primarily involved in:
Degradation of surface receptor proteins
Formation of endocytic multivesicular bodies (MVBs)
Nuclear envelope sealing
Mitotic spindle disassembly during late anaphase
Membrane fission events, including viral budding (notably HIV-1)
The protein exhibits both nuclear and cytoplasmic/vesicular distributions, suggesting multifunctional roles in cellular processes.
Zebrafish CHMP2A shares significant structural homology with human CHMP2A, consistent with the conservation of ESCRT-III components across vertebrates. Functionally, both proteins:
Form part of the core ESCRT-III machinery
Participate in membrane deformation and scission events
Interact with other ESCRT components like CHMP3 to form polymeric structures
The structure of CHMP2A-CHMP3 heterodimers reveals that CHMP2A can fold into the same closed conformation structure as CHMP3, with their hairpin tips shifted by six helical turns relative to each other . This structural conservation suggests functional conservation across species, making zebrafish an appropriate model for studying CHMP2A-related processes relevant to human biology.
Zebrafish (Danio rerio) provides numerous advantages for investigating CHMP2A function:
The zebrafish model bridges the gap between cell culture-based test systems and more complex vertebrate models, offering a cost-effective and efficient system for CHMP2A functional studies .
Based on the available literature, the optimal developmental stages for studying CHMP2A in zebrafish depend on the specific biological process under investigation:
Early embryogenesis (0-24 hpf): For studying CHMP2A's role in fundamental developmental processes. Zygotic genome activation occurs by 16 hpf, making this a critical window for examining early CHMP2A expression .
Larval stages (3-5 dpf): Particularly appropriate for renal function studies, as the pronephros becomes fully functional at 96 hpf . This stage allows for analysis of CHMP2A's role in physiological processes.
Adult stages: For studying complex processes like immune-mediated antitumor activity where mature immune system interactions are required .
Most studies utilize embryonic and early larval stages (24-120 hpf) for CHMP2A research due to their experimental tractability and the establishment of major organ systems. When working with transient knockout approaches, microinjection of CRISPR/Cas9 components should be performed at the one-cell stage for maximum efficiency .
For generating CHMP2A knockout or knockdown models in zebrafish, researchers can employ several approaches:
CRISPR/Cas9-mediated knockout:
Design guide RNAs targeting the zebrafish chmp2a gene
Microinject the Cas9 protein and guide RNA complex into one-cell stage embryos
For transient knockout studies, directly use the mosaic F0 larvae
For stable knockout lines, raise F0 fish to adulthood, cross them, and screen for germline transmission in F1 and F2 generations
Validate knockout efficiency using sequencing and protein expression analysis
Morpholino-based knockdown:
Design antisense morpholinos targeting the translation start site or splice junctions of chmp2a
Microinject morpholinos into one-cell stage embryos
Include appropriate controls (standard control morpholino, rescue experiments)
Validate knockdown efficiency by RT-PCR (for splice-blocking morpholinos) or western blot
For highest experimental rigor, phenotypes should be confirmed using both approaches and rescued by co-injection with wild-type chmp2a mRNA to establish specificity.
When designing experiments to investigate CHMP2A's role in zebrafish development, researchers should consider:
Protocol standardization:
Different experimental parameters can significantly affect results. Data from the NTP DNT-DIVER database showed that when comparing protocols with different parameters, concordance dropped and potency shift was on average about 3.8-fold for cumulative developmental toxicity outcomes .
Critical experimental parameters:
| Parameter | Consideration | Impact |
|---|---|---|
| Fish strain | Different strains (AB, 5D Tropical) may have varying baseline phenotypes | Affects reproducibility across laboratories |
| Chorion status | Dechorionated vs. intact | Affects compound penetration and exposure |
| Exposure timing | Early (6 hpf) vs. late (72 hpf) | Determines developmental processes affected |
| Exposure scenario | Static vs. static renewal | Influences compound stability and effective dose |
| Developmental endpoints | Time point of analysis (96 hpf vs. 120 hpf) | Different developmental processes may be observable |
| Sample size | Adequate embryo numbers | Statistical power to detect subtle phenotypes |
Controls and validation:
Include wild-type controls from the same clutch
Use positive controls with known developmental phenotypes
Validate key findings with secondary approaches (e.g., confirm CRISPR results with morpholinos)
Consider maternal contribution of CHMP2A, which may mask early phenotypes
Zebrafish CHMP2A models offer valuable insights into human disease mechanisms through several approaches:
Cancer research: CHMP2A has been identified as a regulator of immune cell-mediated antitumor activity. Zebrafish models can help elucidate how CHMP2A influences tumor microenvironment and immune evasion mechanisms. Studies have shown that CHMP2A mediates tumor cell resistance to NK cell-mediated cytotoxicity by regulating secretion of extracellular vesicles expressing immune-modulating ligands .
Neurodegenerative disease models: Human CHMP2A has been associated with Frontotemporal Dementia And/Or Amyotrophic Lateral Sclerosis 7 . Zebrafish models can reveal how CHMP2A dysfunction contributes to neurodegeneration through altered membrane trafficking or protein degradation pathways.
Developmental disorders: By studying CHMP2A in zebrafish embryogenesis, researchers can identify critical developmental processes requiring ESCRT-III function and connect these to congenital human disorders.
Kidney disease: The zebrafish pronephros serves as an excellent model for studying renal function. CHMP2A's role in the ESCRT pathway is likely important for kidney cell homeostasis, and zebrafish models can reveal mechanisms relevant to human kidney disorders .
Infectious disease: CHMP2A plays roles in viral budding, including HIV-1. Zebrafish models can help elucidate host-pathogen interactions mediated by ESCRT components .
The conserved nature of ESCRT pathways across vertebrates makes zebrafish findings highly translatable to human disease contexts.
Advanced imaging techniques for tracking CHMP2A dynamics in live zebrafish include:
Fluorescent fusion proteins:
Generation of CHMP2A-GFP/mCherry transgenic lines using Tol2 transgenesis
Conditional expression systems (GAL4/UAS) to control spatio-temporal expression
Photo-convertible fluorescent tags (e.g., Dendra2) to track protein movement
CRISPR-based tagging:
CRISPR knock-in of fluorescent tags at the endogenous chmp2a locus for physiological expression levels
Split-GFP complementation to visualize CHMP2A-partner interactions
Selective plane illumination microscopy (SPIM):
Allows long-term 3D imaging with reduced phototoxicity
Particularly valuable for tracking CHMP2A dynamics during embryogenesis
High-speed confocal microscopy:
For capturing rapid ESCRT-III assembly/disassembly events
Spinning disk systems offer improved temporal resolution
Correlative light and electron microscopy (CLEM):
Combines fluorescence imaging of CHMP2A with ultrastructural analysis
Reveals the relationship between CHMP2A localization and membrane remodeling events
The transparency of zebrafish larvae makes them particularly amenable to these advanced imaging approaches, enabling visualization of labeled CHMP2A at single-cell resolution within the context of developing tissues .
CHMP2A plays significant roles in immune responses, particularly in tumor immune evasion mechanisms. Based on recent studies, the following methodologies are optimal for investigating CHMP2A's immunological functions in zebrafish:
Syngeneic tumor models:
Flow cytometry analysis:
Characterization of tumor-infiltrating immune populations
Quantification of NK cells, T cells, and myeloid-derived suppressor cells
Assessment of activation markers on immune cells
Extracellular vesicle isolation and characterization:
Differential ultracentrifugation of conditioned media
Nanoparticle tracking analysis of EV size distribution
Proteomics analysis of EV cargo to identify immune-modulating molecules
Research has shown that CHMP2A regulates secretion of EVs expressing NK cell activating ligands such as MHC class I chain-related proteins, which act as decoys to inhibit NK cell killing of tumor cells. Targeted deletion of CHMP2A can enhance immune-mediated antitumor activity in vivo .
CHMP2A, as a component of the ESCRT-III complex, likely plays important roles in zebrafish kidney development and function, particularly in the formation and maintenance of the pronephros. Experimental approaches to investigate this include:
Pronephros-specific manipulations:
Targeted knockdown or knockout of chmp2a in pronephric tissues
Use of kidney-specific promoters (wt1a, cdh17) for tissue-specific expression
Functional assessment of pronephros:
The zebrafish larva pronephros is fully functional at 96 hpf and exhibits:
Filtration and clearance assays:
The experimental protocol developed by Huwyler et al. uses intravenous injections of calibrated amounts of fluorescent reference compounds to study renal function in 3-4 dpf larvae. This approach allows for precise dosing and defined exposure while providing information on tolerability, circulation behavior, extravasation, cellular interaction, and tissue accumulation .
| Transporter | Substrate | Inhibitor | Method of Detection |
|---|---|---|---|
| ABC transporters (abcb4, mrp1, mrp2, mrp4) | NBD-CsA, Sulfo101, fluo-cAMP | MK-571, verapamil, erythromycin | Fluorescence in tubular lumen or disappearance from blood |
| SLC transporters (oat/slc22) | Fluorescein | Probenecid, p-aminohippurate | Fluorescence in tubular lumen |
| Folate receptor (distal tubule) | FITC-folate-PEG2000 | - | Fluorescence in tubular cells |
This comprehensive approach enables detailed characterization of CHMP2A's role in renal development and function .
Researchers working with CHMP2A in zebrafish models face several technical challenges:
Maternal contribution masking phenotypes:
CHMP2A protein and mRNA deposited maternally may obscure early developmental phenotypes
Solution: Use maternal-zygotic mutants or maternal protein degradation approaches (e.g., Trim-Away technology)
Functional redundancy:
Other ESCRT-III components may compensate for CHMP2A loss
Solution: Generate combined knockouts of multiple ESCRT components or use dominant-negative approaches
Variability between laboratories:
Different protocol parameters can significantly affect results. Data from the NTP DNT-DIVER database showed that laboratories with similar protocol parameters had concordance as high as 86%, but when protocols differed, concordance dropped significantly .
Solution: Standardize key protocol parameters:
| Parameter | Recommendation |
|---|---|
| Fish strain | Use consistent strain (e.g., AB wild-type) |
| Chorion status | Standardize (dechorionated or intact) across experiments |
| Exposure timing | Consistent exposure window for developmental studies |
| Sample size | Adequately powered (minimum 20-30 embryos per group) |
| Controls | Include within-clutch controls |
Lethality of complete knockout:
Complete CHMP2A loss may cause early embryonic lethality
Solution: Use conditional knockouts, hypomorphic alleles, or time-controlled CRISPR approaches
Distinguishing direct vs. indirect effects:
CHMP2A functions in fundamental cellular processes, making it difficult to separate primary from secondary effects
Solution: Use tissue-specific or inducible approaches to target CHMP2A function with spatial and temporal precision
Cellular localization challenges:
Comprehensive validation of zebrafish CHMP2A models requires multiple complementary approaches:
Genomic validation:
PCR and sequencing to confirm CRISPR-induced mutations
Analysis of indel spectrum and predicted protein consequences
Assessment of potential off-target effects using whole genome sequencing
Transcript analysis:
RT-PCR to assess mRNA levels and potential alternative splicing
RNA-seq to evaluate global transcriptional consequences
In situ hybridization to confirm tissue-specific loss of expression
Protein validation:
Western blotting using validated antibodies against zebrafish CHMP2A
Immunohistochemistry to assess protein expression patterns
Mass spectrometry-based proteomics to confirm protein absence and identify compensatory changes
Functional validation:
Rescue experiments using wild-type CHMP2A mRNA to confirm specificity
Phenotypic comparison with other ESCRT-III component knockouts
Assessment of known CHMP2A-dependent processes (e.g., MVB formation)
Cross-validation with alternative approaches:
Compare CRISPR knockout with morpholino knockdown phenotypes
Use chemical inhibitors of ESCRT-III function as complementary approach
Employ dominant-negative CHMP2A variants as an alternative strategy
Reproducibility assessment:
Thorough validation is essential for distinguishing specific CHMP2A-related phenotypes from off-target effects or general developmental disruptions.
Several cutting-edge technologies are poised to transform zebrafish CHMP2A research:
Base editing and prime editing:
Allows for precise modification of CHMP2A without double-strand breaks
Enables introduction of specific disease-associated mutations
Reduces off-target effects compared to traditional CRISPR/Cas9
Single-cell multi-omics:
Single-cell RNA-seq to map CHMP2A expression across all cell types during development
Single-cell ATAC-seq to identify regulatory elements controlling CHMP2A expression
Spatial transcriptomics to preserve tissue context while analyzing expression patterns
Optogenetic and chemogenetic tools:
Light- or drug-inducible CHMP2A variants for temporal control
Allows for cell-specific and reversible manipulation of CHMP2A function
Enables study of acute vs. chronic CHMP2A perturbation
Advanced microscopy:
Super-resolution imaging to visualize ESCRT-III assembly dynamics
Light-sheet microscopy for long-term, non-toxic imaging of CHMP2A during development
Correlative light and electron microscopy to link CHMP2A localization with ultrastructural features
Tissue-specific CRISPR screens:
In vivo screens to identify genetic interactors of CHMP2A
Cell-type-specific knockout libraries to dissect tissue-specific functions
Pooled screening approaches using single-cell RNA-seq as readout
Zebrafish organoids and explants:
These emerging technologies will enable more precise, dynamic, and comprehensive studies of CHMP2A function in development, disease, and cellular processes.
Comparative studies of CHMP2A across different model organisms provide valuable insights into its conserved and divergent functions:
Evolutionary conservation analysis:
CHMP2A's high conservation across species suggests fundamental cellular roles
Comparing CHMP2A sequences and structural features across vertebrates can identify critical functional domains
Cross-species rescue experiments (e.g., human CHMP2A in zebrafish knockouts) can test functional conservation
Multi-model comparative approaches:
Parallel studies in zebrafish, mice, and cell culture systems can distinguish organism-specific from universal functions
Each model offers complementary advantages:
| Model | Advantage for CHMP2A Research |
|---|---|
| Zebrafish | In vivo visualization, high-throughput screening, vertebrate developmental context |
| Mouse | Mammalian physiology, complex immune system, extensive genetic tools |
| Cell culture | Biochemical studies, high-resolution imaging, precise molecular manipulation |
| Yeast | Fundamental ESCRT mechanisms, genetic screening, structural studies |
Disease-specific comparative studies:
Comparing CHMP2A function in disease contexts across models
Human patient-derived cells alongside zebrafish models carrying equivalent mutations
Validation of zebrafish findings in mammalian systems for translational relevance
Developmental role comparison:
Structural biology integration:
These comparative approaches leverage the strengths of multiple model systems to build a comprehensive understanding of CHMP2A's fundamental functions across evolution.