VPS4B Human

Vacuolar Protein Sorting 4 Homolog B Human Recombinant
Shipped with Ice Packs
In Stock

Description

Gene and Protein Structure

The VPS4B gene spans 3337 base pairs and encodes a 444-amino-acid protein (51.8 kDa) with a conserved AAA-ATPase domain. Key structural features include:

  • N-terminal MIT domain: Binds ESCRT-III substrates.

  • Large and small AAA ATPase domains: Facilitate ATP hydrolysis and hexamer formation.

  • C-terminal helix: Unique structural element involved in membrane interactions .

Table 1: Comparison of Human VPS4 Paralogs

FeatureVPS4AVPS4B
Chromosomal location16q11.218q21.33
Protein identity81% similarity to VPS4B81% similarity to VPS4A
Cellular localizationEndosomes, cytokinetic bridgeEndosomes, cytokinetic bridge
Knockout phenotypeEmbryonic lethal (mouse)Embryonic lethal (mouse)
Disease associationsNeurodevelopmental disordersCancer, CDA
Sources:

Functional Roles in Cellular Processes

VPS4B orchestrates ESCRT-III filament disassembly and membrane fission in:

  • Multivesicular body (MVB) formation: Facilitates degradation of ubiquitinated membrane proteins via lysosomes .

  • Cytokinetic abscission: Remodels ESCRT-III filaments during cell division. Depletion delays abscission, with VPS4A acting earlier and VPS4B later in the process .

  • Viral budding: Essential for HIV-1 release by severing membrane connections .

Cancer

  • Synthetic lethality: Co-depletion of VPS4A and VPS4B induces immunogenic cell death in colorectal cancer (CRC) cells, releasing DAMPs that activate anti-tumor M1 macrophages .

  • Frequent deletions: VPS4B loss occurs in 20–30% of CRC cases, correlating with poor prognosis .

Neurodevelopmental Disorders

  • Homozygous VPS4B knockout causes embryonic lethality in mice, while heterozygous models show no overt defects. In humans, dysregulation is linked to impaired neural pruning and brain structural abnormalities .

ATPase Activity

  • VPS4B forms hexameric or dodecameric complexes stabilized by co-factor VTA1, enhancing ATP hydrolysis efficiency .

  • The central pore loop is critical for ESCRT-III substrate extraction, as mutations here disrupt HIV-1 budding .

Table 2: Key Research Findings

Study FocusModel SystemKey Outcome
Abscission timingHCT116 cellsVPS4A depletion delays abscission > VPS4B
Synthetic lethalityCRC xenograftsDual VPS4A/B silencing inhibits tumor growth
Structural analysisX-ray crystallographyRevealed MIT domain flexibility and β-domain role in co-factor binding
Sources:

Therapeutic Potential

  • Targeted inhibition: Small-molecule disruptors of VPS4B-VTA1 interaction reduce cancer cell migration and invasion .

  • Recombinant protein tools: His-tagged VPS4B (1–444 aa) is used to study ATPase kinetics and ESCRT-III interactions .

Research Challenges and Future Directions

  • Paralog redundancy: Functional overlap with VPS4A complicates mechanistic studies.

  • Druggability: Developing isoform-specific inhibitors remains challenging due to high sequence similarity .

Product Specs

Introduction
Vacuolar protein sorting-associated protein 4B (VPS4B) is a member of the AAA (ATPases associated with diverse cellular activities) family. It plays a role in the final stages of the endosomal multivesicular bodies (MVB) pathway. VPS4B interacts with membrane-bound ESCRT-III complexes and facilitates their disassembly, likely in conjunction with membrane fission. Dominant negative mutations of VPS4B hinder vacuolar protein sorting and also interfere with HIV-1 and MLV budding.
Description
Recombinant human VPS4B, expressed in E. coli, is a single, non-glycosylated polypeptide chain. It consists of 468 amino acids (with amino acids 1-444 present) and has a molecular weight of 51.8kDa. This VPS4B protein is fused to a 24 amino acid His-tag at its N-terminus and is purified using proprietary chromatographic techniques.
Physical Appearance
A clear, colorless solution, sterilized by filtration.
Formulation
The VPS4B protein is supplied in a solution at a concentration of 0.25mg/ml. The solution also contains 20mM Tris-HCl buffer (pH 8.0), 0.15M NaCl, 30% glycerol, and 1mM DTT.
Stability
For short-term storage (up to 2-4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to freeze the product at -20°C. Adding a carrier protein like HSA or BSA (0.1%) is advisable for long-term storage. Repeated freezing and thawing should be avoided.
Purity
Purity is determined to be greater than 90% using SDS-PAGE analysis.
Synonyms
Vacuolar Protein Sorting 4 Homolog B (S. Cerevisiae), SKD1, Suppressor Of K+ Transport Defect 1, Cell Migration-Inducing Gene 1 Protein, Suppressor Of K(+) Transport Growth Defect 1,Vacuolar Protein Sorting 4B (Yeast), SKD1B, PS4-2, Cell Migration-Inducing 1, Vacuolar Protein Sorting 4B, Vacuolar Protein Sorting-Associated Protein 4B, EC 3.6.4.6, VPS42, Protein SKD1.
Source
E.coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSHMSSTSP NLQKAIDLAS KAAQEDKAGN YEEALQLYQH AVQYFLHVVK YEAQGDKAKQ SIRAKCTEYL DRAEKLKEYL KNKEKKAQKP VKEGQPSPAD EKGNDSDGEG ESDDPEKKKL QNQLQGAIVI ERPNVKWSDV AGLEGAKEAL KEAVILPIKF PHLFTGKRTP WRGILLFGPP GTGKSYLAKA VATEANNSTF FSISSSDLVS KWLGESEKLV KNLFQLAREN KPSIIFIDEI DSLCGSRSEN ESEAARRIKT EFLVQMQGVG VDNDGILVLG ATNIPWVLDS AIRRRFEKRI YIPLPEPHAR AAMFKLHLGT TQNSLTEADF RELGRKTDGY SGADISIIVR DALMQPVRKV QSATHFKKVR GPSRADPNHL VDDLLTPCSP GDPGAIEMTW MDVPGDKLLE PVVSMSDMLR SLSNTKPTVN EHDLLKLKKF TEDFGQEG.

Q&A

What is the primary cellular function of VPS4B and how is it regulated in human cells?

VPS4B is an ATPase family protein that plays crucial roles in the endosomal sorting complex required for transport (ESCRT) pathway, specifically in multivesicular body (MVB) formation, virus budding, and cytokinesis abscission . Methodologically, VPS4B function can be studied using:

  • Mutational analysis of ATP-binding domains to understand enzymatic function

  • Fluorescence microscopy with tagged VPS4B to track subcellular localization

  • Co-immunoprecipitation to identify protein interaction partners

  • Serum starvation and refeeding experiments to study its regulation during cell cycle progression, as demonstrated in Huh7 and HepG2 HCC cells where VPS4B expression increases after serum refeeding

The protein functions through ATP-dependent disassembly of ESCRT-III complexes, representing the final step in membrane severing processes critical for endocytic sorting, cytokinesis, and membrane repair . Its activity can be directly measured using the Transcreener ADP2 Assay, which detects ADP produced by VPS4B under various experimental conditions .

How do VPS4A and VPS4B differ structurally and functionally despite their high sequence similarity?

VPS4A and VPS4B are paralogs with 81% sequence identity that likely cooperate by forming hetero-oligomers . Despite their similarity, these proteins show important differences:

  • Genomic location: VPS4A and VPS4B are located on separate chromosomes (16q and 18q, respectively)

  • Expression patterns: In colorectal cancer samples, VPS4B expression is significantly downregulated during progression from adenoma to adenocarcinoma, while VPS4A mRNA levels remain unchanged

  • Functional redundancy: Research indicates that VPS4A and VPS4B are mostly functionally redundant, but their differential expression across tissues suggests tissue-specific roles

To study their functional differences experimentally:

  • Generate paralog-specific antibodies verified through immunohistochemistry staining in normal human tissues with known high/low protein abundance

  • Design paralog-specific siRNAs that can efficiently silence the expression of a single paralog without affecting the other

  • Perform synthetic lethality screens using CRISPR/Cas9 methods to evaluate their interdependence

What methodologies are most effective for studying VPS4B localization and trafficking in live cells?

For studying VPS4B localization and trafficking in live cells, researchers should consider these methodological approaches:

  • Fluorescent protein tagging: Generate VPS4B-GFP fusion constructs, ensuring tag placement doesn't interfere with function (preferably C-terminal tagging)

  • Live-cell confocal microscopy: Track VPS4B movements using spinning disk or lattice light-sheet microscopy for high temporal resolution

  • Photoactivatable or photoconvertible fluorescent proteins: Utilize mEOS or PA-GFP fusions to pulse-chase specific VPS4B populations

  • Co-localization studies: Combine with markers for endosomes (Rab5, Rab7), multivesicular bodies (CD63), and midbodies for cytokinesis studies

  • FRAP (Fluorescence Recovery After Photobleaching): Measure VPS4B dynamics and turnover rates at specific cellular locations

When designing these experiments, researchers should validate that:

  • Tagged constructs retain ATPase activity (using in vitro ATPase assays)

  • Expression levels approximate endogenous VPS4B to avoid artifacts from overexpression

  • Controls include ATP-binding deficient mutants to determine how enzymatic activity affects localization

How does VPS4B expression vary across different cancer types, and what are the methodological considerations for accurate assessment?

VPS4B expression shows striking variability across cancer types, with different patterns observed in distinct cancer contexts:

  • Hepatocellular carcinoma (HCC): VPS4B is significantly upregulated in HCC tissues compared to adjacent nontumorous samples, correlating with multiple clinicopathological factors including AJCC stage, microvascular invasion, Ki-67 expression, and poor prognosis .

  • Colorectal cancer (CRC): VPS4B is significantly downregulated during progression from adenoma to adenocarcinoma, with the VPS4B locus frequently deleted .

  • Multiple cancer types: According to the Dependency Map (DepMap) portal data, VPS4B copy number alterations occur across various cancer cell lines, with certain lines showing decreased VPS4B copy number .

For accurate assessment of VPS4B expression, researchers should employ multiple complementary methods:

MethodAdvantagesLimitationsBest Practices
qRT-PCRQuantitative, high sensitivityDoes not assess protein levelsUse multiple reference genes; validate primers for paralog specificity
Western blotProtein-level assessmentSemi-quantitativeValidate antibody specificity against VPS4B KO controls
ImmunohistochemistrySpatial information, clinical samplesSubjective scoringUse validated antibodies and standardized scoring systems
Copy number analysisGenomic alterationsDoesn't reflect expressionCombine with expression analysis

When reporting VPS4B expression changes, researchers should consistently include proper controls and validate findings through multiple methodologies, as demonstrated in studies that confirmed antibody specificity by staining normal human tissues with known high/low protein abundance of VPS4 paralogs .

What is the relationship between VPS4B expression and patient prognosis in different cancers, and how should researchers interpret contradictory findings?

The relationship between VPS4B expression and patient prognosis appears to be cancer-type dependent, with seemingly contradictory findings:

In hepatocellular carcinoma (HCC):

  • High VPS4B expression correlates with poor prognosis

  • Univariate and multivariate survival analyses demonstrate that VPS4B serves as an independent prognostic factor for survival in HCC patients

  • High VPS4B expression correlates with advanced AJCC stage, microvascular invasion, and increased Ki-67 expression

In colorectal cancer (CRC):

  • VPS4B downregulation is observed during progression from adenoma to adenocarcinoma

  • The VPS4B locus is frequently deleted in CRC

To interpret these seemingly contradictory findings, researchers should:

  • Examine tissue specificity: VPS4B may function differently in different tissue contexts

  • Consider cancer hallmarks: Analyze whether VPS4B affects distinct cancer hallmarks in different tumors (proliferation vs. invasion)

  • Evaluate genetic context: Assess whether VPS4B functions differently depending on co-occurring mutations

  • Standardize methodologies: Use consistent methods for expression analysis and patient stratification

  • Perform functional studies: Go beyond correlation to establish causative relationships through in vitro and in vivo knockout/knockdown studies

When designing clinically relevant studies, researchers should use large cohorts with comprehensive clinical annotations and multivariate analyses that account for confounding factors like treatment history and comorbidities.

What experimental approaches best demonstrate the functional consequences of VPS4B alterations in cancer progression?

To robustly demonstrate the functional consequences of VPS4B alterations in cancer progression, researchers should employ a comprehensive experimental framework:

  • In vitro functional assays:

    • Cell proliferation: Use CCK-8 assay and flow cytometry to assess cell cycle progression

    • Migration and invasion assays: Boyden chamber or wound healing assays

    • Colony formation assays: Evaluate long-term growth potential upon VPS4B knockdown or overexpression

  • Genetic manipulation approaches:

    • CRISPR/Cas9 for complete gene knockout: Verify bi-allelic knockout through Sanger sequencing and immunoblotting

    • siRNA for transient knockdown: Use multiple independent siRNA sequences to control for off-target effects

    • Rescue experiments: Re-express wild-type or mutant VPS4B to confirm specificity

  • In vivo models:

    • Xenograft models: Compare tumor growth kinetics between VPS4B-manipulated and control cells

    • Patient-derived xenografts: Use PDX models from VPS4B-high and VPS4B-low tumors

    • Metastasis assays: Tail vein injection or orthotopic implantation to assess metastatic potential

  • Mechanistic investigations:

    • Transcriptome analysis: RNA-seq to identify gene expression changes upon VPS4B alteration

    • Proteomics: Mass spectrometry to identify altered protein networks

    • ESCRT pathway analysis: Evaluate impacts on MVB formation, receptor degradation, and exosome release

Researchers investigating HCC should note that VPS4B knockdown in Huh7 and HepG2 cells led to cell cycle arrest and reduced cell proliferation, suggesting an oncogenic role . Conversely, in CRC contexts, the relationship may be more complex, potentially involving synthetic lethality mechanisms with VPS4A .

How can researchers effectively design experiments to validate synthetic lethality between VPS4A and VPS4B across different cancer types?

Validating synthetic lethality between VPS4A and VPS4B requires a systematic experimental approach:

  • Cell line selection strategy:

    • Use genetically diverse cell lines from multiple cancer types

    • Include lines with natural VPS4B deficiency (e.g., HOP62, SNU410)

    • Leverage public data resources like the Dependency Map (DepMap) to identify cell lines with predicted VPS4A/B dependencies

  • Genetic manipulation approaches:

    • Individual and combined knockdown using validated siRNAs targeting VPS4A and/or VPS4B

    • CRISPR/Cas9-mediated knockout of VPS4B followed by VPS4A depletion

    • Inducible shRNA systems for temporal control of gene silencing

  • Viability assessment methods:

    • Short-term viability: MTT/MTS/CellTiter-Glo assays

    • Long-term survival: Colony formation assays

    • Cell death mechanism: Annexin V/PI staining, caspase activation assays

  • Critical controls:

    • Non-targeting siRNA controls

    • Single gene knockdowns

    • Rescue experiments with siRNA-resistant constructs

    • Isogenic control cell lines (e.g., HCT116 VPS4B+/+)

  • In vivo validation:

    • Xenograft models with dual knockdown systems

    • Patient-derived xenografts with natural VPS4B deficiency

In previously published work, researchers demonstrated that HOP62 and SNU410 cancer cell lines with naturally decreased VPS4B copy number exhibited significantly suppressed cell viability when VPS4A was silenced . This approach of identifying and using cell lines with natural VPS4B deficiency provides compelling evidence for synthetic lethality across cancer types.

What molecular mechanisms underlie the synthetic lethality between VPS4A and VPS4B, and how can they be experimentally elucidated?

The synthetic lethality between VPS4A and VPS4B likely stems from their collaborative role in critical cellular processes. Experimental approaches to elucidate underlying mechanisms include:

  • Transcriptomic analysis:

    • RNA-seq comparing VPS4A, VPS4B, and VPS4A+B depleted cells revealed that combined silencing induced transcription of 587 genes, far exceeding the effects of individual knockdowns

    • Gene ontology analysis showed upregulation of inflammatory response and programmed cell death pathways

  • Cell death mechanism characterization:

    • Concomitant depletion of VPS4A+B leads to G2/M cell cycle arrest due to impaired cytokinesis

    • Activation of caspase 7 and canonical NF-κB signaling

    • Release of damage-associated molecular patterns (DAMPs) that promote inflammatory responses

  • Cellular phenotype assessment:

    • Endocytic system disruption (measured by transferrin uptake)

    • Altered cellular morphology

    • Defects in membrane remodeling and ESCRT-III disassembly

  • Biochemical characterization:

    • ATP hydrolysis assays to measure enzymatic activity

    • Protein complex formation analysis using BN-PAGE or co-immunoprecipitation

    • Structural studies to understand VPS4A/B hetero-oligomerization

  • Immunological consequences:

    • DAMPs released from VPS4A+B-depleted cells activate macrophages toward the anti-tumor M1 phenotype

    • Analyze cytokine release profiles and immune cell activation markers

These mechanisms suggest that VPS4A and VPS4B function as "highly penetrant interactors" as defined by Ryan et al. (2018), making them promising therapeutic targets . The mechanistic understanding supports that VPS4A is the only paralog that can overtake VPS4B function and vice versa, and their proteins likely form direct physical interactions essential for cell survival .

What drug development strategies are most promising for targeting the VPS4A/B synthetic lethality in VPS4B-deficient cancers?

Developing therapeutics targeting the VPS4A/B synthetic lethality represents a promising precision medicine approach for VPS4B-deficient cancers. The most viable strategies include:

  • Small molecule inhibitor development:

    • Target VPS4A ATPase activity in VPS4B-deficient contexts

    • Use high-throughput screening assays like the Transcreener ADP2 Assay

    • Screen parameters:

      • Z' factor of 0.81 indicates robust assay performance

      • Reaction conditions: 50 mM HEPES (pH 7.5), 10 mM MgCl₂, 50 mM KCl, 5 mM DTT, 0.01% Triton, 37°C

      • ATP concentration: 2 μM (sub-Km)

  • Structure-based drug design:

    • Leverage structural differences between VPS4A and VPS4B despite 81% sequence identity

    • Focus on ATP-binding pockets and protein-protein interaction domains

    • Use fragment-based approaches and virtual screening

  • Synthetic lethality enhancers:

    • Identify compounds that synergize with VPS4A inhibition in VPS4B-deficient cells

    • Screen for modulators of pathways activated by VPS4A/B depletion

  • Delivery strategies:

    • Develop nanoparticle formulations for targeted delivery

    • Explore antibody-drug conjugates for cancer-specific targeting

    • siRNA/antisense oligonucleotide approaches for VPS4A knockdown

  • Biomarker development:

    • VPS4B expression/deletion as companion diagnostic

    • Develop IHC methods for accurate VPS4B protein detection

    • Genomic markers for VPS4B locus deletion (18q)

  • Immunomodulatory potential:

    • Leverage the finding that VPS4A+B depletion induces inflammatory cell death

    • Explore combination with immune checkpoint inhibitors to enhance anti-tumor immunity

When prioritizing these approaches, researchers should consider that VPS4A and VPS4B are "highly penetrant interactors" as their proteins participate in multiple essential pathways across many cell types . This characteristic suggests that VPS4A inhibitors could have a broad therapeutic window in VPS4B-deficient cancers.

What are the optimal conditions and considerations for developing a robust VPS4B activity assay for inhibitor screening?

Developing a robust VPS4B activity assay requires careful optimization of multiple parameters:

  • Enzyme source and preparation:

    • Recombinant human VPS4B (aa 1-444) expressed and purified from E. coli has been successfully used

    • Ensure proper protein folding and activity through quality control testing

    • Consider His-tagged vs. untagged protein for different applications

  • Reaction buffer optimization:

    • Standard conditions: 50 mM HEPES (pH 7.5), 10 mM MgCl₂, 50 mM KCl, 5 mM DTT, 0.01% Triton

    • Systematically test pH ranges (7.0-8.0) and salt concentrations

    • Include reducing agents (DTT or BME) to maintain enzyme stability

    • Add detergent (e.g., 0.01% Triton) to prevent protein aggregation

  • Substrate considerations:

    • ATP concentration: Use sub-Km concentration (e.g., 2 μM) for inhibitor studies

    • Consider including ESCRT-III protein components or peptide substrates

  • Detection method selection:

    • ADP detection using Transcreener ADP2 Assay offers direct measurement of enzymatic activity

    • Fluorescence polarization (FP), fluorescence intensity (FI), or TR-FRET readouts are compatible

    • Ensure stop solution effectively terminates the reaction (200 mM HEPES pH 7.5, 400 mM EDTA, 0.2% Brij-35)

  • Assay validation parameters:

    • Z' factor: Values ≥0.7 indicate excellent assay quality (demonstrated Z' = 0.81)

    • Signal-to-background ratio: Optimize for maximum difference

    • Reaction linearity: Verify under initial velocity conditions

    • DMSO tolerance: Test compound solvent compatibility

  • Plate format considerations:

    • Compatible with 96, 384, and 1536-well formats

    • Use non-binding surface plates for FP/FI measurements (e.g., Corning 384-well plates Cat. #4514)

  • Controls and standards:

    • Include ATP/ADP standard curve for data normalization

    • Positive control: Known VPS4 inhibitors or ATP-competitive inhibitors

    • Negative control: Heat-inactivated enzyme

Following these guidelines will help establish a robust screening platform for identifying potent and selective VPS4B inhibitors with therapeutic potential.

What are the most common challenges in studying VPS4B function in cellular contexts, and how can researchers overcome them?

Researchers studying VPS4B function face several technical challenges that can be addressed through specific methodological approaches:

  • Functional redundancy with VPS4A

    • Challenge: VPS4A can compensate for VPS4B loss, masking phenotypes

    • Solution: Use double knockdown/knockout approaches

    • Approach: Apply validated siRNAs with demonstrated specificity for each paralog

  • Distinguishing VPS4B-specific effects

    • Challenge: Separating VPS4B functions from general ESCRT pathway disruption

    • Solution: Compare phenotypes between VPS4B knockout and other ESCRT component disruptions

    • Approach: Create a panel of ESCRT component knockouts in the same cell background

  • Pleiotropic cellular effects

    • Challenge: VPS4B affects multiple cellular processes (endocytosis, cytokinesis, viral budding)

    • Solution: Use synchronized cells or acute protein depletion

    • Approach: Apply auxin-inducible degron systems for temporal control

  • Antibody specificity issues

    • Challenge: Cross-reactivity between VPS4A and VPS4B due to 81% sequence identity

    • Solution: Validate antibodies against knockout controls

    • Approach: Test antibodies in tissues with known high/low protein abundance and in VPS4B knockout cell lines

  • Technical issues with activity assays

    • Challenge: Establishing physiologically relevant activity measurements

    • Solution: Optimize reaction conditions and substrate concentrations

    • Approach: Use the Transcreener ADP2 Assay with optimized buffer conditions

  • Variable expression across tissues

    • Challenge: Tissue-specific effects and expression patterns

    • Solution: Analyze cell-type specific functions using appropriate models

    • Approach: Compare effects across multiple cell types representing different tissues

  • In vivo model development

    • Challenge: Creating viable animal models due to potential developmental effects

    • Solution: Use conditional or inducible knockout systems

    • Approach: Tissue-specific Cre-loxP systems or inducible shRNA approaches

Researchers have successfully addressed these challenges by using multiple complementary approaches, such as validating antibody specificity through staining in tissues with known expression patterns and in CRISPR/Cas9-engineered VPS4B knockout cell lines .

How should researchers interpret and troubleshoot variability in VPS4B expression and function across different experimental systems?

Variability in VPS4B expression and function across experimental systems requires systematic analysis and troubleshooting:

  • Sources of variability in expression studies:

Variable FactorPotential ImpactTroubleshooting Approach
Cell culture conditionsSerum factors affect VPS4B expression Standardize serum lots and culture protocols
Cell confluencyMay alter expression/localizationMaintain consistent cell density across experiments
Antibody specificityFalse positives/negativesValidate with multiple antibodies and knockout controls
RNA integrityAffects qPCR resultsVerify RNA quality (RIN >8) before analysis
Reference gene selectionImpacts normalizationUse multiple validated reference genes
  • Functional assay variability troubleshooting:

  • Cell cycle effects: VPS4B function varies throughout the cell cycle; synchronize cells when necessary

  • Knockdown efficiency: Verify protein reduction by Western blot alongside functional assays

  • Compensatory mechanisms: Assess VPS4A upregulation in response to VPS4B depletion

  • Genetic background differences: Compare results across multiple cell lines with documented genetic backgrounds

  • Technical variation: Implement robust normalization methods and include appropriate controls

  • Analysis of contradictory findings:

When faced with conflicting results, systematically evaluate:

  • Differences in experimental models (cell lines, primary cells, tissues)

  • Method-specific artifacts (antibody specificity issues, siRNA off-target effects)

  • Biological context (cancer type, genetic background, disease stage)

  • Temporal factors (acute vs. chronic depletion of VPS4B)

  • Standardization recommendations:

  • Generate and share validated reagents (antibodies, constructs, cell lines)

  • Report detailed methodological parameters (reaction conditions, antibody validation)

  • Include appropriate positive and negative controls

  • Validate findings through complementary methodological approaches

For example, researchers investigating VPS4B in colorectal cancer validated antibody specificity by testing in tissues with known expression patterns and in engineered VPS4B knockout cells , while studies on VPS4B enzymatic activity established reproducible conditions (50 mM HEPES pH 7.5, 10 mM MgCl₂, 50 mM KCl, 5 mM DTT, 0.01% Triton) with statistical validation (Z' factor of 0.81) .

How might single-cell technologies advance our understanding of VPS4B function in heterogeneous tumor populations?

Single-cell technologies offer unprecedented opportunities to investigate VPS4B function in heterogeneous tumor contexts:

  • Single-cell RNA sequencing (scRNA-seq) applications:

    • Map VPS4B expression patterns across distinct tumor cell subpopulations

    • Identify correlations between VPS4B expression and cancer stem cell markers

    • Discover cell state-specific dependencies on VPS4B function

    • Track transcriptional consequences of VPS4B perturbation at single-cell resolution

  • Single-cell proteomics approaches:

    • Quantify VPS4B protein abundance across tumor cells using mass cytometry (CyTOF)

    • Employ proximity ligation assays to detect VPS4A/VPS4B interactions in situ

    • Analyze phosphorylation status and other post-translational modifications

  • Spatial transcriptomics integration:

    • Map VPS4B expression to specific tumor microenvironments (hypoxic regions, invasive front)

    • Correlate VPS4B status with immune infiltration patterns

    • Analyze spatial relationships between VPS4B-high and VPS4B-low tumor regions

  • Methodological considerations:

    • Sample preparation must preserve cell viability and prevent stress responses

    • Transcript dropout requires computational correction strategies

    • Validation of key findings using orthogonal methods (e.g., RNAscope, IF)

    • Integration of multiple data modalities using computational approaches

  • Experimental design innovations:

    • CRISPR-based lineage tracing to follow VPS4B-deficient clones during tumor evolution

    • Combined single-cell RNA-seq with functional readouts (e.g., CRISPR screens)

    • Patient-derived models with preserved tumor heterogeneity

This approach will be particularly valuable for understanding the clinical implications of heterogeneous VPS4B expression in cancers where both high and low expression have been reported in different contexts, such as HCC (high expression) versus CRC (frequent deletions) .

What are the implications of VPS4B's role in the ESCRT pathway for emerging fields like extracellular vesicle research and viral pathogenesis?

VPS4B's central role in the ESCRT pathway has significant implications for extracellular vesicle biology and viral pathogenesis:

  • Extracellular vesicle (EV) biogenesis and cargo sorting:

    • VPS4B functions in MVB formation, a key step in exosome biogenesis

    • Altered VPS4B expression may modify EV composition and release rates

    • Research opportunities include:

      • Proteomics and RNA-seq of EVs from VPS4B-manipulated cells

      • Live imaging of EV biogenesis using fluorescently tagged VPS4B

      • Investigation of cancer-specific EV alterations linked to VPS4B status

  • Viral pathogenesis mechanisms:

    • VPS4B participates in virus budding for multiple virus families

    • Methodological approaches:

      • Virus production quantification in VPS4B-depleted cells

      • Structure-function analysis of VPS4B domains involved in viral protein interactions

      • Development of antivirals targeting VPS4B-virus interactions

  • Plasma membrane repair:

    • ESCRT machinery including VPS4B mediates membrane repair

    • Research avenues:

      • Laser wounding assays in VPS4B-deficient cells

      • Analysis of membrane repair kinetics using calcium imaging

      • Investigation of cancer cell survival under mechanical stress

  • Neurodegenerative disease connections:

    • ESCRT dysfunction is implicated in neurodegeneration

    • Study approaches:

      • Analysis of VPS4B expression in neurodegenerative disease tissues

      • Evaluation of protein aggregation in VPS4B-deficient neuronal models

      • Investigation of autophagy-lysosome pathway alterations

  • Methodological innovations needed:

    • Improved techniques for isolating pure EV populations

    • Advanced imaging methods for visualizing ESCRT dynamics at membranes

    • Development of VPS4B activity modulators with paralog specificity

    • Organoid and 3D culture systems to study VPS4B in physiologically relevant contexts

The synthetic lethality between VPS4A and VPS4B suggests redundancy in these critical functions, implying that therapeutic targeting strategies must carefully consider impacts on these essential cellular processes.

How can systems biology approaches integrate VPS4B function within broader cellular networks to identify new therapeutic vulnerabilities?

Systems biology approaches offer powerful frameworks for contextualizing VPS4B function within cellular networks and identifying novel therapeutic vulnerabilities:

  • Multi-omics data integration:

    • Combine transcriptomics, proteomics, and phosphoproteomics data from VPS4B-perturbed systems

    • Analyze data from VPS4A, VPS4B, and VPS4A+B depleted cells to construct differential network models

    • Generate protein interaction networks centered on VPS4B using affinity purification-mass spectrometry

    • Computational approach: Apply weighted gene correlation network analysis (WGCNA) to identify co-expressed gene modules

  • Network perturbation analysis:

    • Map synthetic lethal interactions through systematic genetic screens

    • Use the DepMap and similar datasets to identify context-dependent vulnerabilities

    • Apply computational algorithms to predict synthetic lethal partners beyond VPS4A

    • Experimental validation: Combinatorial CRISPR screens targeting ESCRT and related pathway components

  • Dynamic modeling approaches:

    • Develop mathematical models of ESCRT pathway dynamics incorporating VPS4B function

    • Simulate effects of VPS4B inhibition across different genetic backgrounds

    • Identify network bottlenecks and critical nodes as potential drug targets

    • Validation approach: Time-course experiments with acute VPS4B depletion

  • Therapeutic vulnerability identification:

    • Apply network-based drug repurposing strategies targeting VPS4B-associated pathways

    • Analyze cancer dependency maps to find cancer types with heightened VPS4B dependency

    • Use machine learning to predict drug combinations synergizing with VPS4B inhibition

    • Experimental testing: High-throughput drug combination screens in VPS4B-deficient cells

  • Methodology requirements:

    • Develop computational infrastructure for integrating heterogeneous data types

    • Implement rigorous statistical methods for network inference

    • Establish experimental systems for high-throughput phenotypic validation

    • Design reporter systems for real-time monitoring of VPS4B activity in living cells

Previous research demonstrated the power of this approach by using the Dependency Map portal to identify cell lines vulnerable to VPS4A depletion specifically when VPS4B copy number was decreased . This systems-level analysis led to the selection of HOP62 and SNU410 cell lines for experimental validation of synthetic lethality, exemplifying how computational approaches can guide targeted experimental design.

What are the most promising translational applications of VPS4B research, and what methodological advances are needed to realize these opportunities?

VPS4B research offers several promising translational applications that require specific methodological advances:

  • Prognostic biomarker development:

    • VPS4B expression serves as an independent prognostic factor in HCC

    • VPS4B deletion/downregulation is frequent in CRC

    • Needed advances:

      • Standardized IHC protocols with validated antibodies

      • Integration with existing cancer staging systems

      • Prospective clinical validation studies

  • Therapeutic targeting of synthetic lethality:

    • VPS4A inhibition in VPS4B-deficient cancers represents a precision medicine strategy

    • Required developments:

      • High-throughput screening platforms for VPS4A inhibitors

      • Structure-based drug design leveraging differences between paralogs

      • Delivery systems for cancer-specific targeting

  • Immunotherapeutic applications:

    • VPS4A+B depletion induces immunomodulatory cell death

    • DAMPs released activate macrophages toward anti-tumor M1 phenotype

    • Methodological needs:

      • Characterization of immune response to VPS4-targeting therapies

      • Identification of optimal combination strategies with existing immunotherapies

      • Development of in vivo models with intact immune systems

  • Diagnostic applications:

    • VPS4B status as a companion diagnostic for VPS4A-targeting therapies

    • Technical requirements:

      • Development of clinical-grade assays for VPS4B copy number/expression

      • Establishment of validated cutoff values for patient stratification

      • Integration with existing molecular diagnostic platforms

  • Drug resistance mechanisms:

    • Understanding how cancer cells might develop resistance to VPS4A/B-targeting strategies

    • Methodological approaches:

      • Generation of resistant cell lines through long-term drug exposure

      • Whole-genome CRISPR screens to identify resistance mechanisms

      • Patient-derived models to capture clinical resistance patterns

These translational applications build upon foundational discoveries, such as the synthetic lethality between VPS4A and VPS4B and the prognostic significance of VPS4B in HCC . The field now requires focused investment in methodological advances to bridge the gap between these discoveries and clinical applications.

What experimental design considerations are essential for researchers planning to investigate VPS4B in novel contexts or disease models?

Researchers planning to investigate VPS4B in novel contexts should adopt a comprehensive experimental design framework that accounts for VPS4B's complex biology:

  • Model system selection considerations:

    • Match model to research question (cell lines vs. organoids vs. animal models)

    • Consider VPS4B baseline expression levels and genomic status

    • Account for VPS4A expression and potential compensatory mechanisms

    • Include models spanning normal, pre-malignant, and malignant states when studying disease progression

  • Genetic manipulation strategy:

    • Choose appropriate technology (RNAi vs. CRISPR) based on experimental goals

    • Design proper controls (isogenic cell lines, non-targeting sequences)

    • Consider inducible systems for temporal control of VPS4B depletion

    • Plan for rescue experiments with wild-type and mutant VPS4B

  • Multidimensional phenotypic analysis:

    • Assess cell viability using complementary methods (short-term vs. long-term)

    • Examine cell cycle effects using flow cytometry

    • Evaluate endocytic function through transferrin uptake or similar assays

    • Investigate cellular morphology and ultrastructure changes

  • Molecular characterization depth:

    • Implement multi-omics approaches (transcriptomics, proteomics)

    • Perform pathway analysis to contextualize findings

    • Validate key findings using orthogonal methods

    • Consider single-cell approaches for heterogeneous systems

  • Translational relevance enhancement:

    • Include patient-derived materials when possible

    • Correlate experimental findings with clinical databases

    • Develop assays with potential for clinical implementation

    • Consider impact of standard-of-care treatments on observed phenotypes

  • Reproducibility and rigor elements:

    • Use multiple cell lines or models to ensure generalizability

    • Implement blinding and randomization where appropriate

    • Define physiologically relevant endpoints a priori

    • Plan for appropriate statistical analysis with sufficient power

These design considerations are exemplified in published work combining mechanistic studies, such as RNA-seq and functional assays in VPS4-depleted cells, with clinical correlation using patient samples and tissue microarrays .

How should researchers approach the development of VPS4B-targeting therapeutics in light of its essential cellular functions and potential toxicity concerns?

Developing VPS4B-targeting therapeutics requires careful consideration of selectivity, specificity, and potential toxicity:

  • Target selection strategy:

    • Focus on VPS4A inhibition in VPS4B-deficient contexts rather than direct VPS4B targeting

    • Consider the therapeutic window based on differential expression between normal and cancer tissues

    • Evaluate tissue-specific expression patterns to predict potential toxicities

    • Explore context-dependent vulnerabilities using dependency mapping data

  • Drug modality considerations:

    • Small molecules: Target ATP-binding domain or protein-protein interaction sites

    • Peptide-based inhibitors: Disrupt specific protein interactions

    • Degraders (PROTACs): Achieve selective VPS4A degradation

    • RNA therapeutics: Enable tissue-specific delivery and expression modulation

  • Selectivity optimization approaches:

    • Structure-based design targeting paralog-specific regions

    • Allosteric inhibitors to achieve specificity

    • Fragment-based drug discovery to identify selective chemical matter

    • Phenotypic screening with toxicity counterscreens

  • Preclinical toxicity assessment:

    • Evaluate effects on normal cells with varying VPS4B expression levels

    • Establish in vitro to in vivo toxicity correlation

    • Implement intermittent dosing schedules to mitigate toxicity

    • Develop toxicity biomarkers for early detection of adverse effects

  • Combination strategy exploration:

    • Identify synergistic combinations requiring lower drug concentrations

    • Target compensatory pathways activated upon VPS4A/B inhibition

    • Explore sequential treatment schedules to minimize toxicity

    • Consider immune-modulating combinations leveraging immunogenic cell death

  • Biomarker-driven development:

    • Develop companion diagnostics for VPS4B status

    • Identify pharmacodynamic markers of target engagement

    • Establish predictive biomarkers of response

    • Monitor resistance mechanisms through longitudinal sampling

This development approach is supported by evidence that VPS4A is a highly penetrant synthetic lethal partner for VPS4B , suggesting that selective targeting may have a therapeutic window in appropriate genetic contexts.

Product Science Overview

Gene and Protein Structure

The VPS4B gene is located on chromosome 18 at band 18q21.33 . The gene encodes a protein that consists of 444 amino acids . VPS4B shares a high degree of amino acid sequence similarity with its paralog, VPS4A, and with the yeast Vps4 and mouse Skd1 proteins .

Function

VPS4B plays a crucial role in the late steps of the endosomal multivesicular bodies (MVB) pathway. It recognizes membrane-associated ESCRT-III assemblies and catalyzes their ATP-dependent disassembly, possibly in combination with membrane fission . This process is essential for the redistribution of ESCRT-III components to the cytoplasm for further rounds of MVB sorting .

Biological Processes

VPS4B is involved in various biological processes, including:

  • Endosomal microautophagy
  • Regulation of centrosome duplication
  • Protein depolymerization
  • Viral budding via host ESCRT complex
  • Cholesterol transport
  • Negative regulation of exosomal secretion
  • Multivesicular body assembly
  • Regulation of mitotic spindle assembly
  • Cell division
  • Endosomal transport
  • Response to lipid
  • Ubiquitin-independent protein catabolic process via the multivesicular body sorting pathway
  • Positive regulation of G2/M transition of mitotic cell cycle
  • Positive regulation of exosomal secretion
  • Septum digestion after cytokinesis
  • Mitotic metaphase plate congression
  • Endosome to lysosome transport via multivesicular body sorting pathway
  • Cell cycle
  • ESCRT III complex disassembly
  • Macroautophagy
  • Transport
  • Positive regulation of viral process
  • Vacuole organization
  • Midbody abscission .
Clinical Significance

Mutations or dysregulation of the VPS4B gene have been associated with various diseases, including Dentin Dysplasia, Type I . The protein’s involvement in critical cellular processes makes it a significant target for research in understanding and potentially treating these conditions.

Research and Applications

Human recombinant VPS4B is used in various research applications to study its function and role in cellular processes. It is particularly valuable in understanding the mechanisms of intracellular protein trafficking and the endosomal sorting pathway.

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 2024 Thebiotek. All Rights Reserved.