Charged multivesicular body protein 5 (CHMP5) plays a crucial role in multivesicular body (MVB) biogenesis and the downregulation of signaling pathways through receptor degradation . MVBs are late endosomes that are essential in the internalization of nutrients, ligands, and receptors via the endolysosomal system . Thus, MVBs are critical for sorting membrane proteins for degradation or routing to the lysosome . CHMP5 is also involved in programmed cell death, antiviral mechanisms, the maintenance of centrosomes, and cellular cytokinesis .
Xenopus tropicalis has emerged as a model for studying human disease genes because of its diploid genome, which shows a high degree of synteny to humans, and its relatively short life cycle .
T-cell receptor (TCR) stimulation affects CHMP5-knockdown (KD) cells, suggesting CHMP5 is implicated in TCR-induced activation of NF-κB .
| Experiment | Result |
|---|---|
| PMA treatment (100 ng/mL) | Marginal increases in p65- and p50-DNA-binding activities |
| CHMP5 KD Jurkat cells compared to control cells | Markedly higher p65- and p50-DNA-binding activities, suggesting CHMP5 deficiency is implicated in NF-κB activation |
CHMP5 serves as an anti-apoptotic protein and may participate in leukemogenesis .
Apoptosis: After CHMP5 was inhibited, caspase 3 was activated in CHMP5-deficient U937 cells, which indicates the apoptotic pathway was activated . CHMP5 silencing activated the Granzyme B/Perforin apoptotic pathway .
Necrosis: The apoptosis-inducing protein-mediated necrotic PCD pathway is activated after CHMP5 inhibition .
Autophagy: CHMP5 inhibition results in an increase in Bcl-2, a proposed inhibitor of autophagy . Autophagic PCD did not occur in CHMP5-deficient U937 cells .
After CHMP5 inhibition, the Granzyme B/Perforin apoptotic pathway and AIF-mediated caspase-independent necrosis pathway are activated .
Charged multivesicular body protein 5 (CHMP5) is likely a peripherally associated component of the endosomal sorting complex required for transport III (ESCRT-III). It plays a role 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, including stimulated growth factor receptors, lysosomal enzymes, and lipids.
KEGG: xtr:549502
UniGene: Str.20965
CHMP5 (Charged multivesicular body protein 5) is a component of the endosomal sorting required for transport complex III (ESCRT-III) involved in multivesicular bodies (MVBs) formation and sorting of endosomal cargo proteins. It participates in the invagination and scission processes that generate intraluminal vesicles (ILVs) for lysosomal degradation of membrane proteins, growth factor receptors, and lipids .
Xenopus tropicalis offers several distinct advantages as a model organism:
Diploid genome (unlike the allotetraploid X. laevis), simplifying genetic studies
High degree of synteny with mammalian genomes, often in stretches of hundreds of genes
Well-established CRISPR/Cas9 protocols for efficient gene editing
Ability to generate thousands of embryos per day via natural mating or in vitro fertilization
Unilateral mutation technique (injecting only one cell at 2-cell stage) providing within-animal controls
These advantages make X. tropicalis particularly suitable for high-throughput, parallelized analysis of gene function relevant to human disease mechanisms .
Recombinant X. tropicalis CHMP5 (219 amino acids, 24.8 kDa) can be produced using several expression systems:
Expression hosts:
E. coli (most common for basic structural studies)
Yeast systems
Baculovirus-infected insect cells
Standard E. coli expression protocol:
Clone the full CHMP5 coding sequence into an appropriate expression vector (pGEX for GST-tag or pET for His-tag)
Transform into an expression strain (BL21(DE3) or similar)
Grow cultures at 37°C to OD600 of 0.5-0.6
Induce with IPTG (typically 0.2-0.5 mM)
Shift to lower temperature (16-18°C) for overnight expression
Harvest cells and lyse using appropriate buffer systems
Purify using affinity chromatography based on the fusion tag
Consider additional purification steps (ion exchange, size exclusion)
Verify purity by SDS-PAGE and identity by Western blot or mass spectrometry
For Xenopus-specific applications, researchers may adapt protocols from established methods for purifying proteins from Xenopus egg extracts, which can yield more physiologically relevant protein forms with appropriate post-translational modifications .
X. tropicalis CHMP5 has the following key characteristics:
Sequence information:
219 amino acids in length
Molecular weight of 24.8 kDa
Full sequence: MNRLFGKSKPKVPPSTLTDCISNVDSRSESIDKKISRLDAELVKYKDQMKKMREGPSKNMVKQKALRVLKQKRMYEQQRDNLNQQSFNMEQTNYAIQSLKDTKTTVDAMKVGAKEMKKAYKQVKIDQIEDLQDQLEDMMENANEIQEALSRSYGTPEIDEDDLEAELDALGDELLLDDDTSYLDEAASAPAIPEGVPNDSKNKDGVLVDEFGLPQIPAT
Structural features:
Contains coiled-coil domains characteristic of ESCRT-III components
Features a putative bipartite nuclear localization signal (NLS) in the N-terminus, which is present in jawed vertebrates but absent in invertebrate eukaryotes
Conservation:
CHMP5 is highly conserved across vertebrate species
The X. tropicalis sequence shares significant homology with human CHMP5 (also known as Vps60, CGI-34, PNAS-2, or SNF7DC2)
The conservation of sequence and function makes X. tropicalis CHMP5 relevant for understanding human CHMP5 biology
Several sophisticated techniques can be employed to investigate CHMP5 function:
Genetic manipulation:
CRISPR/Cas9 genome editing for knockout or knock-in studies
Unilateral CRISPR injection at 2-cell stage creates embryos with one wild-type side and one mutant side for internal control
Morpholino antisense oligonucleotides for transient knockdown
mRNA overexpression for gain-of-function studies
Protein analysis:
Immunoisolation using cryolysis for Xenopus tissues (limiting yolk protein contamination)
Co-immunoprecipitation to identify interaction partners
Western blotting for expression analysis
Chromatin immunoprecipitation (ChIP) to study nuclear functions
Cellular analysis:
Immunofluorescence microscopy for protein localization
Live imaging with fluorescently tagged proteins
Functional assays:
Endosomal sorting and receptor trafficking assays
Phenotypic analysis of embryonic development in CHMP5-depleted embryos
These methods can be combined to build a comprehensive understanding of both cytoplasmic and nuclear CHMP5 functions.
CHMP5 deficiency has profound effects on endosomal morphology and function, as demonstrated in studies of CHMP5 knockout cells:
Morphological effects:
Enlarged late endosomal compartments
MVBs become abnormally enlarged and heavily packed with internal vesicles
Molecular markers:
Structures positive for CI-M6PR (cation-independent mannose-6-phosphate receptor)
Positive for LBPA (lysobisphosphatidic acid) and LAMP1 (lysosomal-associated membrane protein 1)
More pronounced colocalization of these markers compared to wild-type cells
Functional consequences:
Severely reduced capacity to degrade internalized materials
After a 1-hour pulse of HRP (horseradish peroxidase), wild-type cells degraded nearly all internalized protein after 9 hours, while CHMP5-deficient cells showed minimal degradation
Immunogold labeling revealed approximately five-fold more internalized HRP accumulated within enlarged MVBs in CHMP5-deficient cells
These findings indicate that while CHMP5 is not required for MVB formation itself, it is essential for the normal function of late endosomes and lysosomes in protein degradation.
Studies in mouse models provide insights into the developmental roles of CHMP5 that are likely relevant to Xenopus:
Developmental phenotypes in CHMP5-deficient mice:
Early embryonic lethality around embryonic day 10 (E10)
Severe developmental abnormalities in the ventral region after E7.5
Abnormal neural tube formation
Defects in allantois-chorion fusion
Impaired somite segmentation
Normal embryonic axes initially, followed by severe disorganization
Comparative phenotypes between CHMP5-/- and Hrs-/- mice:
| CHMP5-/- | Hrs-/- |
|---|---|
| Early embryonic lethality around E10 | Similar timing of lethality |
| Most mutant embryos smaller than wild-types at E7.5 | Similar size defects |
| Defect in ventral folding morphogenesis | Present |
| Cardia bifida and massive cell death in ventral region around E8.5 | Present |
| No fusion of allantois with chorion | Present |
| No somite segmentation | Present |
| Enlarged M6PR and LAMP1-positive endosomal compartments | Enlarged TfR-positive endosomal compartments |
| Enlarged MVBs result from heavily packed internal vesicles | Enlarged MVBs result from defect of vesicular invagination |
This comparison highlights the specific nature of CHMP5 function in MVB formation compared to other ESCRT components .
CHMP5 plays critical roles in receptor trafficking that impact multiple signaling pathways:
Receptor trafficking effects:
Regulates turnover and down-regulation of receptors, including TGF-β receptors
CHMP5 depletion leads to accumulation of receptors in endosomal compartments
Affects receptor recycling versus degradation decisions
Influences duration and intensity of receptor-mediated signaling
Impact on signaling pathways:
TGF-β signaling: CHMP5 deficiency affects TGF-β receptor turnover
Notch signaling: Potentially affects Notch receptor processing
Growth factor signaling: Likely impacts signaling via receptor tyrosine kinases
Experimental approaches to study these functions:
Receptor internalization and degradation assays
Signaling reporter assays to measure pathway activation
Biochemical analysis of receptor levels and modifications
Genetic interaction studies between CHMP5 and pathway components
Understanding these functions is critical as receptor trafficking defects underlie many developmental disorders and diseases.
Recent studies have revealed unexpected nuclear roles for CHMP5 in gene regulation:
Nuclear functions of CHMP5:
Associates with BRD4 (Bromodomain-containing protein 4) on chromatin
Binds to specific gene loci, including enhancers and promoters
Promotes H3K27 acetylation at enhancers and super-enhancers
Experimental approaches:
Nuclear localization studies:
Create NLS-deletion mutants by removing the N-terminal nuclear localization signal
Compare cellular distribution of wild-type versus ΔNLS-CHMP5 by immunofluorescence
Perform nuclear/cytoplasmic fractionation followed by Western blotting
Chromatin association:
Protein-protein interactions:
Functional consequences:
These approaches can distinguish nuclear functions from cytoplasmic ESCRT-related roles.
CHMP5 depletion produces distinct cellular phenotypes depending on the cell type and context:
In T-ALL cells (CUTLL1):
Proliferation defect with cell cycle arrest at S phase
Impaired G2/M progression
Significant gene expression changes (1057 upregulated and 702 downregulated genes)
Downregulation of MYC and other critical T-ALL genes
Impaired RNA polymerase II pause release
In embryonic stem (ES) cells:
Normal fluid phase endocytosis
Greatly reduced capacity to degrade internalized material
Accumulation of cargo within enlarged MVBs
Approximately five-fold more internalized HRP accumulated compared to wild-type cells
In embryonic cells:
Enlarged endosomal compartments positive for late endosomal markers
Abnormal colocalization of endosomal and lysosomal markers
Defective receptor degradation
These phenotypes highlight the dual roles of CHMP5 in endosomal function and gene regulation.
While CHMP5's core functions are conserved, there are important species-specific considerations:
Similarities across species:
Basic ESCRT-III component functions in MVB formation
Role in receptor trafficking and degradation
Involvement in embryonic development
Unique aspects in Xenopus tropicalis:
Diploid genome simplifies genetic studies compared to X. laevis
High conservation with mammalian CHMP5 including the N-terminal NLS
Developmental context offers insights into vertebrate-specific functions
X. tropicalis has distinct experimental advantages for embryological studies
Comparative limitations:
Some specialized structures in mammals (like the placenta) are absent in Xenopus
Differences in immune system development and function
Aquatic versus terrestrial adaptations
Complementary nature of different models:
Mouse models provide mammalian-specific insights but are more costly
Cell culture models offer mechanistic detail but lack developmental context
Xenopus offers a middle ground with vertebrate relevance and experimental accessibility
Understanding these differences helps researchers select the appropriate model system and interpret results in the context of human biology.
Isolating CHMP5 protein complexes from Xenopus tissues requires specialized protocols:
Sample preparation using cryolysis:
Flash-freeze Xenopus tissues or embryos in liquid nitrogen
Cryogenically grind samples using a mortar and pestle kept at liquid nitrogen temperature
This approach limits contamination from abundant yolk proteins while preserving native protein complexes
Immunoprecipitation protocol:
Extract proteins using a gentle lysis buffer containing appropriate protease inhibitors
Pre-clear lysates with protein A/G beads to reduce non-specific binding
Incubate with anti-CHMP5 antibodies or antibodies against epitope tags if using tagged CHMP5
Capture complexes using magnetic beads conjugated with appropriate secondary antibodies
Wash thoroughly while maintaining conditions that preserve protein-protein interactions
Elute complexes under native or denaturing conditions depending on downstream applications
Analysis methods:
Western blotting to confirm presence of CHMP5 and suspected interaction partners
Mass spectrometry for unbiased identification of complex components
Functional reconstitution assays to test activity of isolated complexes
This methodology can be extended for proteomic analysis of CHMP5-associated complexes in different developmental contexts or subcellular compartments .
CRISPR/Cas9 mutagenesis in X. tropicalis provides powerful approaches to study CHMP5 function:
Comprehensive protocol:
Design stage:
Design 2-3 sgRNAs targeting early exons of the CHMP5 gene
Verify target specificity using Xenbase (https://www.xenbase.org) resources
Consider targeting conserved functional domains
sgRNA and Cas9 preparation:
Synthesize sgRNAs using in vitro transcription
Prepare Cas9 protein or mRNA (protein often gives higher efficiency)
Microinjection:
For complete knockout: inject both cells at 2-cell stage
For unilateral knockout (preferred): inject only one cell at 2-cell stage to create within-animal control
Typical injection mix: 500 pg Cas9 protein + 300 pg sgRNA
Validation:
Collect samples at early stages to verify editing efficiency
Use T7 endonuclease assay or direct sequencing of PCR products
Verify protein loss by Western blot if antibodies are available
Phenotype analysis:
Examine morphological development at appropriate stages
Compare injected versus uninjected sides in unilateral injections
Perform molecular and cellular analyses to characterize effects
The unilateral CRISPR approach is particularly valuable as it allows direct comparison between wild-type and mutant tissues within the same embryo, controlling for any variation between individuals .
CHMP5 has distinctive roles within the ESCRT-III complex compared to other components:
Unique aspects of CHMP5 function:
Acts as a peripherally associated component of ESCRT-III
CHMP5 deficiency results in enlarged MVBs with abundant internal vesicles, unlike other ESCRT-III mutants which typically show defects in MVB formation
This phenotypic difference suggests CHMP5 functions after internal vesicle formation, potentially in cargo sorting or processing
Comparative functional analysis:
| Function | CHMP5 | Other ESCRT-III components |
|---|---|---|
| MVB formation | Not essential for vesicle formation | Core components (CHMP4) are essential |
| Internal vesicle formation | MVBs contain abundant internal vesicles in CHMP5-/- | Deletion typically blocks internal vesicle formation |
| Degradative capacity | Required for efficient degradation | Similar requirement |
| Nuclear functions | Has NLS and associates with BRD4 | Most lack nuclear functions |
| Embryonic requirement | Essential (lethality at E10 in mice) | Variable depending on component |
Hierarchical assembly:
ESCRT-III complex assembly is sequential
CHMP5 appears to function at late stages of the process
May be involved in ESCRT-III disassembly or recycling
Understanding these functional differences is crucial for interpreting phenotypes and designing targeted experiments to study specific aspects of MVB biology.
Recent studies reveal that CHMP5 has unexpected nuclear roles in gene regulation:
Mechanisms of CHMP5-mediated gene regulation:
Association with chromatin regulators:
Effects on transcriptional machinery:
Target genes and pathways:
Disease relevance:
Promotes T-cell acute lymphoblastic leukemia (T-ALL) by controlling gene expression programs
May contribute to other cancers through effects on receptor signaling and transcription
Developmental disorders could potentially arise from CHMP5 dysfunction
These findings reveal CHMP5 as a dual-function protein with both cytoplasmic ESCRT-related roles and nuclear gene regulatory functions that impact development and disease.
Investigating post-translational modifications (PTMs) of CHMP5 requires specialized techniques:
Sample preparation considerations:
Rapid tissue processing to preserve labile PTMs
Use of appropriate inhibitors (phosphatase inhibitors, deubiquitinase inhibitors)
Extraction buffers optimized to maintain native modifications
Analytical techniques:
Mass spectrometry-based approaches:
Enrichment of modified peptides (e.g., TiO2 for phosphopeptides)
Tandem mass spectrometry to identify specific modification sites
Quantitative proteomics to compare modification levels between conditions
Biochemical methods:
Phos-tag SDS-PAGE for phosphorylation analysis
2D gel electrophoresis to separate modified protein forms
Western blotting with modification-specific antibodies
Functional analysis:
Site-directed mutagenesis of modified residues
Phosphomimetic mutations (S/T to D/E) or phospho-null mutations (S/T to A)
Structure-function studies comparing wild-type and mutant proteins
Xenopus-specific considerations: