Recombinant Danio rerio Ubiquitin carboxyl-terminal hydrolase 44 (usp44), partial

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference in order notes for customized preparation.
Lead Time
Delivery times vary depending on the purchasing method and location. Please consult your local distributor for precise delivery estimates.
Note: All proteins are shipped with standard blue ice packs. Dry ice shipping requires advance notification 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. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our standard glycerol concentration is 50% and can serve as a guideline.
Shelf Life
Shelf life depends on various 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
Upon receipt, store at -20°C/-80°C. Aliquoting is essential for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during the manufacturing process.
The tag type is finalized during production. If you require a specific tag, please inform us, and we will prioritize its development.
Synonyms
usp44; zgc:55661; Ubiquitin carboxyl-terminal hydrolase 44; EC 3.4.19.12; Deubiquitinating enzyme 44; Ubiquitin thioesterase 44; Ubiquitin-specific-processing protease 44
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Protein Length
Partial
Purity
>85% (SDS-PAGE)
Species
Danio rerio (Zebrafish) (Brachydanio rerio)
Target Names
usp44
Uniprot No.

Target Background

Function

Deubiquitinase crucial for the spindle assembly checkpoint, preventing premature anaphase onset. It achieves this by specifically deubiquitinating cdc20, a negative regulator of the anaphase-promoting complex/cyclosome (APC/C).

Database Links
Protein Families
Peptidase C19 family, USP44 subfamily
Subcellular Location
Nucleus.

Q&A

What is the function of Ubiquitin carboxyl-terminal hydrolase 44 (usp44) in Danio rerio?

Ubiquitin carboxyl-terminal hydrolase 44 (usp44) belongs to the ubiquitin-specific protease (USP) class of deubiquitylating enzymes (DUBs) in zebrafish. DUBs are essential for modulating protein-protein interactions and signaling processes by removing ubiquitin modifications from substrate proteins. Based on genome-wide loss-of-function studies, many zebrafish DUBs, including those in the USP class, play critical roles in embryonic development, particularly affecting notochord formation . While the specific function of usp44 is not detailed in the search results, DUBs in zebrafish generally regulate important developmental pathways, including Notch and BMP signaling .

How is USP44 structurally characterized compared to other USP family members?

USP44 contains the characteristic UCH (Ubiquitin C-terminal Hydrolase) domain (PF0043) that defines the USP class of deubiquitylating enzymes. The zebrafish genome encodes approximately 51 proteins in the USP class, all sharing this conserved domain . The UCH domain contains the catalytic site responsible for cleaving the isopeptide bond between ubiquitin and substrate proteins. The complete structural characterization of zebrafish USP44 would require techniques such as X-ray crystallography or cryo-electron microscopy, which would reveal its three-dimensional structure and potential substrate-binding regions.

What developmental processes in zebrafish have been linked to USP44 activity?

While the search results don't specifically identify USP44-regulated processes, genome-wide screens have shown that 67% of tested DUBs (57 out of 85) in zebrafish produce observable developmental phenotypes when knocked down . These phenotypes commonly affect multiple regions including head, brain, eyes, body axis, notochord, precardial region, yolk, and tail development. The notochord was consistently affected in all DUB morphants showing phenotypes, suggesting that USP44 likely plays a role in notochord development as well . Further targeted studies would be needed to elucidate USP44's specific developmental functions.

What expression systems are most effective for producing recombinant zebrafish USP44?

Based on successful approaches with other DUBs, methylotrophic yeast Pichia pastoris represents an excellent expression system for recombinant DUBs from various species. For the related DmUCH from Drosophila, P. pastoris yielded high expression levels (up to 210 mg/l) when using the α-mating factor secretion signal . For zebrafish USP44, similar strategies could be employed:

  • Clone the usp44 gene into a suitable P. pastoris expression vector (e.g., pPICZα)

  • Transform the construct into P. pastoris strains (Mut+ or Muts; note that Muts strain produced higher yields for DmUCH)

  • Induce expression using methanol in BMMY medium at optimal pH (pH 6.0 for DmUCH)

  • Harvest the secreted protein from the culture supernatant

Alternative expression systems like E. coli could also be considered, though proper folding of complex eukaryotic proteins may be challenging .

What purification strategy yields the highest activity for recombinant USP44?

A highly effective purification strategy for recombinant USP44 would involve:

  • C-terminal His-tagging of the recombinant protein to facilitate purification

  • Ni-NTA affinity chromatography as the primary purification step

  • Collection and concentration of eluted fractions containing USP44

  • Further purification using size exclusion chromatography or ion exchange chromatography if needed

For the related DmUCH, this approach yielded 18 mg of pure, active enzyme from 100 ml culture broth . Activity testing should be performed at each purification stage using a suitable substrate to ensure the retained enzymatic function. It's important to note that purification conditions should be optimized specifically for USP44 to maintain its native conformation and catalytic activity.

How can the enzymatic activity of purified recombinant USP44 be quantitatively assessed?

To quantitatively assess USP44 enzymatic activity, several methodologies can be employed:

  • Ubiquitin fusion protein cleavage assay: Create a fusion protein with ubiquitin linked to a reporter protein. Incubate with purified USP44 and monitor the release of the reporter protein over time using:

    • SDS-PAGE and Coomassie staining to visualize cleavage products

    • Western blotting for more sensitive detection

    • Mass spectrometry (ESI-MS) to confirm precise cleavage at the C-terminus of ubiquitin

  • Fluorogenic substrate assay: Use commercially available fluorogenic ubiquitin substrates (e.g., Ub-AMC) that release a fluorescent molecule upon cleavage.

  • Ubiquitin-chain disassembly assay: Monitor the disassembly of polyubiquitin chains of various linkage types (K48, K63, etc.) using western blotting with ubiquitin-specific antibodies.

Time-course experiments should be performed to determine reaction kinetics and enzyme efficiency . For the related DmUCH, complete substrate cleavage was observed within 60 minutes .

What morpholino design strategies are most effective for USP44 knockdown in zebrafish embryos?

For effective USP44 knockdown in zebrafish embryos, two primary morpholino (MO) design strategies can be employed:

  • Translation-blocking MO: Target the ATG translation start site of usp44 mRNA to prevent protein synthesis. This approach requires knowledge of the 5' UTR and start codon sequence.

  • Splice-blocking MO: Target exon-intron junctions, particularly those near or within the conserved UCH domain, to disrupt proper splicing. This approach is particularly useful when the complete 5' sequence is unavailable.

Regardless of the strategy, consider these recommendations:

  • Design 2-3 different MOs to control for off-target effects

  • Use 3-5 ng of MO per embryo as a starting concentration

  • Include appropriate controls (standard control MO and/or mRNA rescue)

  • Confirm knockdown efficiency via RT-PCR (for splice-blocking MOs) or western blotting

In the comprehensive zebrafish DUB study, 85 DUBs were successfully targeted using these approaches, with splice-blocking MOs used for 11 genes lacking clear ATG site information .

How do phenotypes of USP44 morphants compare with other DUB family knockdowns in zebrafish?

While the specific phenotype of USP44 morphants isn't detailed in the search results, comprehensive screening of 85 DUB gene knockdowns revealed distinct phenotypic patterns that might be relevant:

  • 67% of tested DUBs (57 out of 85) showed observable developmental phenotypes

  • Most affected multiple regions including head, brain, eyes, body axis, notochord, precardial region, yolk, and tail

  • The notochord was consistently affected in all DUB morphants showing phenotypes

  • Based on neuronal marker (huC) expression patterns, DUBs were classified into five groups (I-V)

To properly compare USP44 with other DUBs, researchers should:

  • Document comprehensive morphological abnormalities at multiple developmental stages

  • Analyze huC expression patterns to determine which group USP44 belongs to

  • Investigate effects on key developmental pathways (Notch, BMP) that other DUB groups regulate

  • Consider genetic interaction studies with related DUBs to identify functional redundancy

What CRISPR-Cas9 strategies are most efficient for generating stable usp44 mutant zebrafish lines?

For generating stable usp44 mutant zebrafish lines using CRISPR-Cas9, consider the following comprehensive strategy:

  • gRNA design:

    • Target the conserved catalytic domain (UCH domain) to ensure loss of enzymatic function

    • Design 2-3 gRNAs targeting different exons within this domain

    • Use algorithms that predict high on-target and low off-target efficiency

    • Avoid regions with SNPs or high variability between strains

  • Delivery method:

    • Microinject a mixture of Cas9 protein (not mRNA) with gRNAs into one-cell stage embryos

    • Recommended concentrations: 300-500 ng/μl Cas9 protein and 25-50 ng/μl of each gRNA

  • Mutation screening:

    • Use T7 endonuclease assay or heteroduplex mobility assay to identify founders with mutations

    • Sequence the target region in F0 mosaic fish to characterize indels

    • Select frameshift mutations that disrupt the catalytic residues

  • Establishing stable lines:

    • Outcross F0 founders to wild-type fish

    • Screen F1 offspring for germline transmission using fin clip genotyping

    • Intercross heterozygous F1 fish to generate homozygous F2 mutants

  • Validation:

    • Confirm loss of USP44 protein using western blotting

    • Perform rescue experiments by injecting wild-type usp44 mRNA

    • Compare phenotypes with morpholino knockdown results to identify potential compensatory mechanisms

This approach should yield stable mutant lines suitable for detailed phenotypic analysis and functional studies.

What methodologies are most effective for identifying physiological substrates of zebrafish USP44?

To identify physiological substrates of zebrafish USP44, several complementary approaches should be employed:

  • Proteomics-based approaches:

    • Stable Isotope Labeling with Amino acids in Cell culture (SILAC) comparing wild-type versus usp44 knockout/knockdown

    • Di-Gly remnant profiling to identify ubiquitinated proteins that accumulate in USP44-deficient cells

    • Proximity-dependent biotin identification (BioID) or APEX2 proximity labeling with USP44 as the bait

  • Co-immunoprecipitation studies:

    • Use catalytically inactive USP44 mutant (trap mutant) that binds but cannot release substrates

    • Perform pull-downs followed by mass spectrometry analysis

    • Validate interactions with candidate substrates using reciprocal co-IPs

  • Yeast two-hybrid screening:

    • Use full-length USP44 or its catalytic domain as bait

    • Screen against zebrafish cDNA libraries from relevant developmental stages

  • In vitro deubiquitination assays:

    • Test candidate substrates identified from above approaches

    • Reconstitute ubiquitination/deubiquitination reactions with purified components

    • Analyze ubiquitin chain types and specificity

  • Comparative analysis:

    • Leverage known substrates of USP44 from other species, particularly mammals

    • Test conservation of these interactions in zebrafish model

These approaches should be complemented with genetic interaction studies and pathway analyses to build a comprehensive picture of USP44's functional network.

How does USP44 activity intersect with major developmental signaling pathways in zebrafish?

While the search results don't specifically address USP44's interaction with developmental pathways, we can make informed inferences based on the general patterns observed with DUBs in zebrafish:

  • Notch signaling pathway:

    • Group I DUBs (otud7b, uchl3, and bap1) have been shown to affect Notch signaling, as evidenced by neuronal hyperplasia phenotypes

    • If USP44 knockdown results in similar neural phenotypes, it may also regulate Notch pathway components

  • BMP signaling pathway:

    • Group IV DUBs (otud4, usp5, usp15, and usp25) play critical roles in dorsoventral patterning through the BMP pathway

    • Assess USP44 morphants for dorsoventral patterning defects indicative of BMP dysregulation

  • Wnt signaling:

    • Many DUBs regulate Wnt pathway components

    • Analyze USP44 morphants for phenotypes consistent with Wnt dysregulation (e.g., tail defects, brain patterning abnormalities)

  • FGF signaling:

    • Examine effects on mesoderm induction and patterning

To determine specific pathway intersections, researchers should:

  • Analyze expression of pathway-specific target genes in USP44-deficient embryos

  • Perform epistasis experiments combining USP44 manipulation with pathway modulators

  • Identify direct USP44 substrates within these pathways using the methodologies described in 4.1

What bioinformatic approaches can predict the ubiquitin chain linkage specificity of zebrafish USP44?

To predict the ubiquitin chain linkage specificity of zebrafish USP44, several bioinformatic approaches can be employed:

  • Structural modeling and analysis:

    • Generate homology models of zebrafish USP44 based on crystal structures of related USP family members

    • Perform molecular docking simulations with differently linked di-ubiquitin molecules (K48, K63, K11, etc.)

    • Analyze the binding pocket and potential specificity-determining residues

  • Sequence-based predictions:

    • Identify conserved ubiquitin-binding motifs and ubiquitin-interacting motifs (UIMs)

    • Compare with USPs of known linkage preferences

    • Analyze the S1' site that determines specificity for the proximal ubiquitin

  • Evolutionary conservation analysis:

    • Compare zebrafish USP44 with homologs from other species where linkage specificity is known

    • Identify conserved residues that may determine specificity

    • Perform phylogenetic analysis to identify closely related DUBs with known specificity

  • Machine learning approaches:

    • Use existing datasets on DUB specificity to train models

    • Apply trained models to predict zebrafish USP44 preferences

    • Validate predictions experimentally

  • Structure-guided mutagenesis predictions:

    • Identify residues likely involved in determining chain specificity

    • Predict effects of mutations on specificity

    • Design experimental validation studies

These computational predictions should be followed by experimental validation using in vitro deubiquitination assays with different ubiquitin chain types.

How conserved is USP44 function between zebrafish and mammals?

The functional conservation of USP44 between zebrafish and mammals can be assessed through multiple approaches:

A robust experimental approach would be to perform cross-species rescue experiments, expressing human USP44 in zebrafish usp44 morphants or mutants to assess functional complementation, which would definitively demonstrate the degree of functional conservation.

What are the evolutionary implications of differences in USP44 structure and function across vertebrate species?

The evolutionary implications of differences in USP44 across vertebrates reveal important insights about deubiquitinating enzyme adaptation:

  • Functional diversification:

    • While the core catalytic function is likely conserved, species-specific adaptations may have evolved in:

      • Substrate recognition domains

      • Regulatory regions that control activity, localization, or stability

      • Interaction interfaces with species-specific partners

  • Expression pattern evolution:

    • Changes in gene regulatory elements may have led to:

      • Different temporal expression during development

      • Tissue-specific expression patterns

      • Responsiveness to different signaling pathways

  • Paralog compensation:

    • The zebrafish genome contains approximately 51 USP-class DUBs

    • Genome duplication events in teleost fish may have created redundancy

    • This may allow for more specialized functions in individual paralogs

    • Some paralogs may compensate for each other, potentially masking phenotypes

  • Selective pressures:

    • Comparative genomic analysis may reveal:

      • Regions under positive selection (potentially involved in species-specific functions)

      • Highly conserved regions (critical for core functions)

      • Rapidly evolving regions (potentially involved in adaptation)

To fully understand these evolutionary implications, researchers should perform comprehensive phylogenetic analyses across vertebrate lineages, focusing on both sequence conservation and functional studies in multiple model organisms.

How can cross-species conservation analysis inform the design of targeted USP44 inhibitors?

Cross-species conservation analysis can significantly inform the design of targeted USP44 inhibitors through several strategic approaches:

  • Identification of conserved catalytic residues:

    • Highly conserved residues in the catalytic domain across species likely represent essential functional sites

    • These conserved sites may serve as primary targets for inhibitor design

    • The catalytic triad of cysteine, histidine, and aspartate residues is particularly important

  • Species-specific binding pocket variations:

    • Structural comparison of USP44 across species can reveal:

      • Conserved binding pocket architecture suitable for broad-spectrum inhibitors

      • Species-specific variations that could be exploited for selective targeting

      • Allosteric sites that may differ between species

  • Differential inhibitor sensitivity prediction:

    • Analyze residues that differ between human and zebrafish USP44

    • Predict how these differences might affect inhibitor binding

    • Design inhibitors that selectively target human USP44 over zebrafish USP44 (or vice versa)

  • Zebrafish as a model for inhibitor testing:

    • Conservation data can inform which aspects of human USP44 function can be reliably modeled in zebrafish

    • Identify conserved pathways affected by USP44 inhibition

    • Predict potential off-target effects based on conservation with other DUBs

  • Evolutionary rate analysis:

    • Regions evolving under purifying selection (slow evolution) represent functionally critical sites

    • Targeting these highly conserved regions may reduce the likelihood of resistance mutations

This approach requires generating accurate structural models of USP44 from multiple species and performing detailed comparative analyses to identify both conserved and divergent features relevant to inhibitor design.

What are the critical parameters for optimizing zebrafish USP44 expression in heterologous systems?

For optimizing zebrafish USP44 expression in heterologous systems, several critical parameters must be carefully controlled:

  • Expression system selection:

    • Pichia pastoris: Demonstrated excellent results for DmUCH with yields up to 210 mg/l

    • Consider strain selection: Mut^s strain produced significantly higher yields than Mut^+ for DmUCH

    • E. coli: May require optimization of codon usage and culture conditions

    • Mammalian cells: Consider for complex post-translational modifications

  • Vector design considerations:

    • Secretion signal: α-mating factor secretion signal worked well for DmUCH in P. pastoris

    • Fusion tags: C-terminal His-tag facilitated purification without affecting activity for DmUCH

    • Promoter selection: For P. pastoris, the AOX1 promoter with methanol induction is recommended

  • Expression conditions optimization:

    • pH optimization: pH 6.0 was optimal for DmUCH expression

    • Temperature: Lower temperatures (20-25°C) may improve folding

    • Induction protocol: For P. pastoris, methanol concentration and feeding schedule

    • Culture medium: BMMY/methanol medium worked well for DmUCH

  • Protein solubility enhancement:

    • Consider fusion partners known to enhance solubility (e.g., SUMO, MBP)

    • Optimize buffer conditions during purification

    • Add stabilizing agents if needed (glycerol, reducing agents)

  • Activity preservation:

    • Validate enzymatic activity at each step of optimization

    • Test various buffer compositions for storage

    • Determine optimal pH and temperature for activity

A systematic approach testing these parameters in factorial designs will help identify the optimal conditions for high-yield, active USP44 production.

What considerations are most important when designing enzymatic assays to distinguish USP44 activity from other DUBs in zebrafish?

Designing enzymatic assays that specifically distinguish USP44 activity from other DUBs in zebrafish requires careful consideration of several factors:

  • Substrate specificity:

    • Develop substrates based on known or predicted USP44-specific targets

    • Design ubiquitin-fusion constructs with peptides derived from these targets

    • Test various ubiquitin chain linkages (K48, K63, K11, etc.) to identify USP44 preferences

  • Assay conditions optimization:

    • Determine pH optima (USPs typically have different pH preferences)

    • Test buffer compositions (salt concentration, reducing agents)

    • Optimize temperature (may reveal USP44-specific temperature sensitivity)

  • Inhibitor profiling:

    • Use available DUB inhibitors with known specificity profiles

    • Develop a fingerprint of inhibitor sensitivity for USP44

    • Include closely related USPs as controls

  • Kinetic parameters determination:

    • Measure K_m and k_cat values for various substrates

    • Compare with other DUBs to identify unique kinetic signatures

    • Analyze reaction progress curves for distinctive patterns

  • Activity in complex samples:

    • Develop immunodepletion strategies to remove USP44 from zebrafish lysates

    • Use activity-based probes that can be coupled with immunoprecipitation

    • Perform differential analysis between wild-type and usp44 knockout samples

A particularly effective approach would be to develop an activity-based probe specifically designed to label active USP44, similar to the HA-tagged ubiquitin-vinylsulfone probes used for other DUBs, but with modifications that enhance selectivity for USP44.

What are the best approaches for developing specific antibodies against zebrafish USP44 for immunological studies?

Developing specific antibodies against zebrafish USP44 requires strategic planning and multiple technical approaches:

  • Antigen design strategy:

    • Full-length protein: Ideal but challenging due to size and potential solubility issues

    • Catalytic domain: More conserved, may cross-react with other USPs

    • Unique epitopes: Target regions specific to USP44 (not conserved in other USPs)

    • Peptide antigens: Select 2-3 unique peptides from non-conserved regions

    • Recombinant fragments: Express soluble fragments containing unique regions

  • Host animal selection:

    • For polyclonal antibodies: Rabbits offer good yields and affinity

    • For monoclonal antibodies: Mouse or rat hybridoma technology

    • Consider chickens for generating antibodies against highly conserved mammalian proteins

  • Antibody validation strategy:

    • Specificity testing:

      • Western blot comparing wild-type vs. usp44 knockout/knockdown samples

      • Immunoprecipitation followed by mass spectrometry

      • Immunohistochemistry with appropriate controls

    • Cross-reactivity assessment:

      • Test against recombinant USP family members

      • Pre-absorption controls with immunizing antigen

  • Purification techniques:

    • Affinity purification against the immunizing antigen

    • Cross-adsorption against related proteins to remove cross-reactive antibodies

    • Consider separate purification of antibody classes for different applications

  • Application-specific validation:

    • Western blot: Test under reducing and non-reducing conditions

    • Immunohistochemistry: Optimize fixation and retrieval methods

    • ChIP applications: Validate for chromatin immunoprecipitation if nuclear roles are anticipated

The most robust approach would involve generating multiple antibodies using different strategies, then extensively validating each for specificity in zebrafish tissues using appropriate genetic controls (morphants or CRISPR mutants).

How can zebrafish USP44 studies inform therapeutic approaches for human diseases linked to deubiquitinating enzyme dysfunction?

Zebrafish USP44 studies can significantly inform therapeutic approaches for human diseases linked to DUB dysfunction through several translational pathways:

  • Disease modeling and drug screening:

    • Generate zebrafish models with mutations corresponding to human disease variants

    • Perform high-throughput chemical screens using these models

    • Assess phenotypic rescue by candidate compounds

    • Filter for compounds with acceptable toxicity profiles

  • Pathway elucidation:

    • Identify conserved pathways regulated by USP44 in both zebrafish and humans

    • Map how USP44 dysfunction impacts these pathways

    • Discover potential alternative intervention points when USP44 function is compromised

    • Genome-wide analysis has revealed zebrafish DUBs involved in Notch and BMP signaling pathways , which are implicated in numerous human diseases

  • Biomarker identification:

    • Characterize molecular signatures associated with USP44 dysfunction

    • Identify conserved biomarkers that could be translated to human diagnostics

    • Develop assays to monitor these biomarkers in patient samples

  • Therapeutic target validation:

    • Validate whether USP44 inhibition or enhancement ameliorates specific disease phenotypes

    • Use genetic approaches (morpholinos, CRISPR) to modulate USP44 activity

    • Test the effects of modulating USP44 at different developmental time points

  • Precision medicine applications:

    • Determine how genetic background influences USP44-related phenotypes

    • Identify genetic modifiers that could predict therapeutic response

    • Develop personalized therapeutic approaches based on genetic profiles

Given that 67% of tested DUBs in zebrafish showed developmental phenotypes when knocked down , zebrafish models offer valuable insights into how DUB dysfunction contributes to human developmental disorders and other diseases.

What insights from zebrafish USP44 research are applicable to understanding cancer mechanisms involving deubiquitylating enzymes?

Zebrafish USP44 research provides valuable insights into cancer mechanisms involving deubiquitylating enzymes through several key areas:

  • Cell cycle regulation:

    • USP44 has been implicated in mitotic checkpoint regulation in mammals

    • Zebrafish models can elucidate how USP44 dysregulation affects cell cycle progression

    • Time-lapse imaging of zebrafish embryos allows real-time visualization of mitotic abnormalities

    • Connect aberrant cell division patterns to cancer-like phenotypes

  • Developmental signaling pathway dysregulation:

    • The genome-wide DUB screen in zebrafish revealed connections to Notch and BMP pathways

    • Both pathways are frequently dysregulated in human cancers

    • Zebrafish studies can reveal how USP44 modulates these pathways and how its dysfunction contributes to tumorigenesis

  • Genetic interaction networks:

    • Screen for genetic interactions between usp44 and known oncogenes/tumor suppressors in zebrafish

    • Identify synthetic lethal interactions that could be targeted therapeutically

    • Map the complete interactome of USP44 substrates relevant to cancer biology

  • Cancer model development:

    • Generate zebrafish lines with usp44 mutations combined with cancer-driving mutations

    • Assess tumor initiation, progression, and metastasis in these models

    • Test potential therapeutic approaches in vivo

  • Drug discovery and validation:

    • Screen small molecule libraries for compounds that modulate USP44 activity

    • Validate hits in zebrafish cancer models

    • Assess specificity profiles and potential off-target effects

    • Determine effects on tumor growth, invasion, and angiogenesis

The comprehensive approach of studying USP44 in zebrafish development establishes a foundation for understanding how its dysregulation contributes to cancer, potentially revealing novel therapeutic targets and strategies.

How can structural information from zebrafish USP44 contribute to structure-based drug design for DUB inhibitors?

Structural information from zebrafish USP44 can significantly advance structure-based drug design for DUB inhibitors through several methodological approaches:

  • Comparative structural analysis:

    • Zebrafish USP44 belongs to the USP class of DUBs containing the characteristic UCH domain (PF0043)

    • Generate homology models based on crystal structures of related USPs

    • Compare active site architecture between zebrafish and human USP44

    • Identify conserved binding pockets suitable for inhibitor development

  • Active site mapping and druggability assessment:

    • Analyze the catalytic triad and surrounding residues

    • Map substrate-binding regions and specificity-determining features

    • Identify allosteric sites that could be targeted

    • Assess druggability of potential binding pockets using computational tools

  • Structure-activity relationship studies:

    • Express and purify recombinant zebrafish USP44 using methods similar to those successful for DmUCH

    • Test candidate inhibitors against purified protein

    • Correlate structural features with inhibitory potency

    • Optimize lead compounds based on structural insights

  • In vivo validation in zebrafish models:

    • Test inhibitor effects on zebrafish embryo development

    • Compare phenotypes with genetic knockdown/knockout models

    • Assess specificity by comparing effects on other DUB knockout models

    • Evaluate toxicity profiles and off-target effects

  • Structural dynamics and molecular simulation:

    • Perform molecular dynamics simulations of zebrafish USP44

    • Identify conformational changes that could be exploited for inhibitor design

    • Compare binding modes of candidate inhibitors

    • Develop computational models to predict inhibitor efficacy

The high expression levels achieved for related DUBs in P. pastoris (210 mg/l for DmUCH) suggest that structural studies of zebrafish USP44 are feasible, potentially providing valuable insights for structure-based drug design that could be translated to human USP44 inhibitors.

What emerging technologies could advance our understanding of USP44 function in zebrafish development?

Several cutting-edge technologies hold promise for advancing our understanding of USP44 function in zebrafish development:

  • CRISPR-based technologies:

    • CRISPR activation/inhibition: CRISPRa/CRISPRi for temporal and tissue-specific modulation of usp44 expression

    • Base editing: Introduce specific point mutations in catalytic residues without double-strand breaks

    • Prime editing: Perform precise edits to create disease-relevant mutations

    • CRISPR screening: Identify genetic interactions with usp44 through multiplexed approaches

  • Advanced imaging techniques:

    • Lattice light-sheet microscopy: Track USP44 dynamics at subcellular resolution in living embryos

    • Expansion microscopy: Visualize protein interactions at nanoscale resolution

    • Correlative light and electron microscopy (CLEM): Connect ultrastructural changes with USP44 function

    • Intravital imaging: Monitor long-term developmental processes in usp44 mutants

  • Single-cell technologies:

    • Single-cell RNA-seq: Map transcriptional changes in individual cells upon usp44 manipulation

    • Single-cell proteomics: Detect cell type-specific changes in protein abundance

    • Spatial transcriptomics: Preserve spatial information while analyzing gene expression changes

    • CITE-seq: Simultaneously profile surface proteins and transcriptomes in developing tissues

  • Proteomic approaches:

    • Proximity labeling: BioID or APEX2 to identify USP44 interactors in vivo

    • Ubiquitinome analysis: Di-Gly remnant profiling to map USP44 substrates

    • Targeted degradation: dTAG or PROTAC approaches for acute USP44 depletion

    • Cross-linking mass spectrometry: Capture transient USP44-substrate interactions

  • Optogenetic and chemogenetic tools:

    • Light-inducible USP44 activity: Spatiotemporal control of enzymatic function

    • Chemically-induced proximity: Rapidly redirect USP44 to specific subcellular locations

    • Degron systems: Control USP44 stability with temporal precision

These technologies will enable researchers to move beyond the traditional morpholino knockdown approaches used in previous zebrafish DUB studies , providing unprecedented insights into USP44 function during development.

What experimental designs would best address the potential redundancy between USP44 and other DUBs in zebrafish?

To address potential redundancy between USP44 and other DUBs in zebrafish, several strategic experimental designs should be considered:

  • Combinatorial genetic knockout approaches:

    • Generate single, double, and triple knockouts of USP44 and closely related DUBs

    • Use CRISPR-Cas9 to efficiently create multiple gene knockouts

    • Analyze phenotypic severity to identify synergistic interactions

    • The comprehensive list of 91 DUBs identified in zebrafish provides candidates for these studies

  • Transcriptional compensation analysis:

    • Compare transcriptional responses to genetic knockout versus morpholino knockdown

    • Identify upregulated DUBs that may compensate for USP44 loss

    • Perform time-course analysis to track compensatory changes

    • Target compensatory DUBs with secondary knockdowns

  • Domain-focused interaction studies:

    • Identify DUBs with similar domain architecture to USP44

    • Generate chimeric proteins swapping domains between USP44 and related DUBs

    • Test whether specific domains from other DUBs can rescue USP44 deficiency

    • Map the structural basis for functional overlap

  • Substrate competition assays:

    • Develop assays to measure deubiquitylation of USP44 substrates

    • Test which other DUBs can process these substrates in vitro

    • Perform substrate trapping in vivo using catalytically inactive mutants

    • Quantify binding affinities and enzymatic efficiencies

  • Evolutionary analysis with functional validation:

    • Identify DUB paralogs resulting from genome duplication in teleost fish

    • Compare with single orthologs in non-duplicated genomes

    • Test functional conservation through cross-species rescue experiments

    • Examine expression patterns for evidence of subfunctionalization

These approaches will build upon the finding that 67% of tested DUBs in zebrafish show developmental phenotypes , suggesting both unique and overlapping functions within this large enzyme family.

What collaborative research frameworks could accelerate zebrafish USP44 research and its translational applications?

To accelerate zebrafish USP44 research and its translational applications, several collaborative research frameworks could be established:

  • Integrated multi-institution zebrafish DUB consortium:

    • Build upon the comprehensive zebrafish DUB phenotype screen

    • Establish standardized protocols for DUB studies in zebrafish

    • Create a centralized repository for zebrafish DUB mutant lines

    • Implement uniform phenotyping guidelines and data submission formats

    • Develop a shared database of DUB phenotypes, interactions, and substrates

  • Cross-species DUB research network:

    • Connect zebrafish USP44 researchers with those studying USP44 in other models

    • Standardize assays to allow direct comparison of results across species

    • Coordinate parallel studies in multiple organisms to identify conserved functions

    • Establish pipelines for rapid translation of zebrafish findings to mammalian models

  • Industry-academia partnerships for drug discovery:

    • Leverage pharmaceutical expertise in DUB inhibitor development

    • Utilize academic strengths in zebrafish disease modeling

    • Establish high-throughput screening facilities optimized for zebrafish assays

    • Create shared intellectual property frameworks to incentivize collaboration

  • Clinical research integration:

    • Connect zebrafish researchers with clinicians studying USP44-related disorders

    • Develop zebrafish models of patient-specific mutations

    • Establish biobanks linking patient samples with corresponding zebrafish models

    • Design clinical trials informed by zebrafish drug screening results

  • Open science infrastructure:

    • Implement pre-registration of zebrafish USP44 studies

    • Establish data sharing platforms for raw experimental data

    • Create open protocols.io collections for standardized methods

    • Develop cloud-based analysis pipelines for phenotypic data

By implementing these collaborative frameworks, the research community can build upon the foundation established by genome-wide screens and expression studies to accelerate both basic understanding of USP44 function and its translation to clinical applications.

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.