Recombinant Dictyostelium discoideum Uncharacterized protein DDB_G0274935 (DDB_G0274935)

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 fulfillment.
Lead Time
Delivery times vary depending on the purchasing method and location. Please contact your local distributor for precise delivery estimates.
Note: Standard shipping includes blue ice packs. Dry ice shipping requires prior arrangement and incurs additional charges.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to collect the contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting at -20°C/-80°C. Our standard glycerol concentration is 50%, serving as a guideline for your use.
Shelf Life
Shelf life depends on various factors: storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized forms have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquot for multiple uses to prevent repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing.
The tag type is determined during production. If you require a specific tag, please inform us, and we will prioritize its development.
Synonyms
DDB_G0274935; Uncharacterized protein DDB_G0274935
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-237
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Dictyostelium discoideum (Slime mold)
Target Names
DDB_G0274935
Target Protein Sequence
MNQHQLHQQL QQQLLQQQQQ QLQYQQQQHN MQQQQQLLQQ QQLKMNIQQQ QQQQQQQQQQ QQQQQQQQQQ QQQLQQQQSP KKTIIQKYNE DLDDMVIKTY TTRLVLTWIY NNPTPPILSP LPKLKVIPNS SSSKQFSNKT LHTSTHTFQD IESYVKEVFD LWDTVLSFFY SQHIKRSKAY YQNVKQSPQN VESDHTVKLT LKSILIGSIY YSDIFFRKSS VLKPKSNFTI LDIVISR
Uniprot No.

Q&A

What makes Dictyostelium discoideum a valuable model organism for studying uncharacterized proteins?

Dictyostelium discoideum serves as an exceptional model organism for several reasons that benefit protein characterization studies. This amoeba has been extensively used to investigate numerous facets of eukaryotic cell biology, including cell motility, cell adhesion, macropinocytosis, phagocytosis, host-pathogen interactions, and multicellular development . Its genome is fully sequenced, and its relatively simple growth requirements make it amenable to laboratory cultivation.

The methodological advantages of D. discoideum for uncharacterized protein studies include:

  • Well-established genetic manipulation techniques

  • Haploid genome facilitating functional genetic studies

  • Conservation of many cellular pathways found in higher eukaryotes

  • Unique life cycle transitions between unicellular and multicellular states

  • Ability to form fruiting bodies, allowing developmental biology studies

These characteristics allow researchers to examine protein function in various cellular contexts that would be difficult to reproduce in other model systems. Furthermore, the relatively small size of the Dictyostelium research community has prompted efforts to develop specialized tools like recombinant antibodies specifically designed for this organism .

How can I approach the initial analysis of an uncharacterized protein like DDB_G0274935?

Methodological approach for initial characterization:

  • Sequence analysis and domain prediction

    • Begin with comparative sequence analysis using BLAST against model organisms

    • Predict protein domains and motifs using tools like SMART, Pfam, and InterPro

    • Identify potential post-translational modification sites

  • Secondary structure analysis

    • Utilize algorithms to predict alpha-helices, beta-sheets, and disordered regions

    • Compare structural predictions with known protein families

  • Cellular localization prediction

    • Use localization prediction software (e.g., PSORT, TargetP)

    • Look for signal peptides or transmembrane domains

  • Ortholog identification

    • Identify orthologs in other Dictyostelium species and other organisms

    • Examine conservation patterns to identify functionally important regions

  • Pathway context

    • Search for information about potential interacting partners

    • Examine expression data to identify co-expressed genes

Following this systematic approach establishes a solid foundation for experimental work and generates testable hypotheses about protein function. For uncharacterized proteins listed in resources like PubChem, additional biochemical pathway information may be available that can guide further investigation .

What experimental design principles should I consider when studying an uncharacterized protein in Dictyostelium?

When designing experiments to characterize DDB_G0274935 or similar uncharacterized proteins, follow these methodological guidelines:

  • Define clear variables

    • Identify your independent variables (e.g., protein expression levels, environmental conditions)

    • Determine appropriate dependent variables (e.g., phenotypic changes, biochemical measurements)

    • Control for extraneous variables that might influence your results

  • Formulate specific hypotheses

    • Develop testable hypotheses based on preliminary sequence analysis

    • Design experiments with appropriate controls to test each hypothesis

  • Consider experimental treatments

    • Plan systematic manipulation of independent variables

    • Determine appropriate sample sizes for statistical validity

    • Implement randomization to minimize bias

  • Select appropriate experimental approaches

    • Choose between between-subjects or within-subjects designs based on your research question

    • Consider true experimental designs with control and experimental groups when possible

  • Plan for measurement consistency

    • Establish reliable methods to measure dependent variables

    • Ensure measurements are reproducible and precise

Experimental Design TypeAdvantagesDisadvantagesSuitability for Uncharacterized Proteins
True Experimental DesignHigh internal validity, establishes causalityResource intensiveIdeal for functional characterization
Quasi-Experimental DesignMore flexible, applicable when randomization is not possibleLower internal validityUseful for initial phenotypic screening
Within-Subjects DesignRequires fewer subjects, controls for individual differencesPotential carryover effectsGood for comparing wild-type and mutant versions
Factorial DesignTests multiple variables simultaneouslyComplex analysisValuable for identifying interaction effects

For rigorous characterization, consider implementing multiple complementary experimental designs to overcome the limitations of any single approach .

How can I optimize recombinant expression of DDB_G0274935 for functional studies?

Optimizing recombinant expression requires methodical troubleshooting and adjustment of multiple parameters:

  • Expression system selection

    • Bacterial systems (E. coli): Fast and economical, but may lack post-translational modifications

    • Eukaryotic systems (insect cells, yeast): Better for complex Dictyostelium proteins

    • Homologous expression in D. discoideum: Optimal for native folding and modifications

  • Expression vector optimization

    • Select promoters appropriate for expression level (constitutive vs. inducible)

    • Include affinity tags for purification (His-tag, GST, etc.)

    • Consider codon optimization for the expression host

  • Culture condition optimization

    • Adjust temperature, pH, media composition, and induction timing

    • Test different cell densities at induction

    • Optimize protein extraction methods based on subcellular localization

  • Purification strategy

    • Implement multi-step purification for higher purity

    • Test different buffer conditions to maintain protein stability

    • Verify protein identity via mass spectrometry

The availability of commercial recombinant Dictyostelium proteins demonstrates the feasibility of successful expression and purification strategies for this organism's proteins .

What CRISPR/Cas9 approaches are most effective for studying uncharacterized proteins in Dictyostelium?

CRISPR/Cas9 genome editing provides powerful tools for functional characterization of uncharacterized proteins in Dictyostelium. The methodological approach includes:

  • sgRNA design and selection

    • Use specialized tools like Cas-Designer or CRISPOR to identify target sites

    • Select sgRNAs with minimal off-target effects

    • Consider the PAM requirements of your Cas9 variant

  • Vector construction

    • Implement Golden Gate assembly for cloning sgRNAs into CRISPR/Cas9 vectors

    • Verify correct integration via colony PCR and Sanger sequencing

    • Select appropriate selection markers for Dictyostelium transformation

  • Transformation protocol

    • Use electroporation for transformation of Dictyostelium cells

    • Apply transient antibiotic selection to induce CRISPR/Cas9 expression

    • Optimize cell recovery conditions post-transformation

  • Mutant screening and validation

    • Implement PCR-based screening to identify knockout mutants

    • Verify mutations through genomic sequencing

    • Confirm phenotypic changes through functional assays

The detailed step-by-step protocol for CRISPR/Cas9-based gene manipulation in Dictyostelium includes:

  • Construction of the CRISPR/Cas9 vector

  • Transformation of Dictyostelium cells

  • Screening for successful knockouts

  • Validation through sequencing and functional analysis

This approach allows for precise genome editing, enabling the generation of knockouts, knockins, and point mutations to study the function of uncharacterized proteins like DDB_G0274935.

What strategies are effective for determining the cellular localization of uncharacterized proteins in Dictyostelium?

Determining cellular localization provides critical insights into protein function. For uncharacterized proteins like DDB_G0274935, employ these methodological approaches:

  • Fluorescent protein tagging

    • Generate C- and N-terminal GFP/RFP fusion constructs

    • Express under native or inducible promoters

    • Validate function is maintained with the tag

    • Image using confocal microscopy with appropriate controls

  • Immunofluorescence microscopy

    • Use antibodies against the target protein or an epitope tag

    • Implement fixation protocols optimized for structure preservation

    • Include markers for different cellular compartments

    • Apply super-resolution techniques for detailed localization

  • Subcellular fractionation

    • Separate cellular components through differential centrifugation

    • Analyze fractions by Western blotting

    • Compare distribution with known compartment markers

    • Quantify relative abundance across fractions

  • Proximity labeling approaches

    • Fuse protein to BioID or APEX2 enzyme

    • Identify neighboring proteins through biotinylation

    • Map protein to known cellular structures

  • Correlative light and electron microscopy

    • Combine fluorescence with ultrastructural analysis

    • Precisely localize proteins at nanometer resolution

These complementary approaches provide robust evidence for cellular localization, helping to develop hypotheses about protein function based on its subcellular context.

How can I determine if my uncharacterized protein participates in known cellular pathways in Dictyostelium?

To integrate an uncharacterized protein into known cellular pathways, implement these methodological strategies:

  • Co-immunoprecipitation and mass spectrometry

    • Express tagged versions of the uncharacterized protein

    • Identify interacting partners through pull-down experiments

    • Validate interactions using reciprocal co-immunoprecipitation

    • Map interactions to known pathways

  • Phenotypic analysis

    • Generate knockout or knockdown strains

    • Systematically test for phenotypes associated with specific pathways

    • Perform rescue experiments with defined mutants

    • Compare phenotypes with known pathway mutants

  • Transcriptomic analysis

    • Compare gene expression profiles between wild-type and mutant strains

    • Identify differentially expressed genes and enriched pathways

    • Look for correlations with known pathway perturbations

    • Validate key findings with RT-qPCR

  • Biochemical assays

    • Test for specific enzymatic activities if sequence suggests catalytic function

    • Measure changes in relevant metabolites or signaling molecules

    • Assess participation in known complexes using size exclusion chromatography

  • Genetic interaction studies

    • Create double mutants with known pathway components

    • Look for synthetic lethality or suppression

    • Perform epistasis analysis to position the protein in a pathway

Resources like PubChem provide information about protein interactions and potential pathway associations that can guide these investigations . For instance, an uncharacterized protein in Dictyostelium (Q54IY4) has been associated with cell cycle pathways, cell cycle checkpoints, and DNA damage response pathways , demonstrating how uncharacterized proteins can be linked to cellular functions.

What are common challenges when working with recombinant Dictyostelium proteins and how can they be addressed?

Researchers frequently encounter several challenges when working with recombinant Dictyostelium proteins, which can be systematically addressed through these methodological approaches:

  • Low expression levels

    • Challenge: Dictyostelium proteins often express poorly in heterologous systems

    • Solution: Optimize codon usage for the expression host, test multiple expression systems (bacterial, insect, yeast), and evaluate different fusion tags (MBP, SUMO) to enhance solubility

  • Protein insolubility

    • Challenge: Formation of inclusion bodies, particularly in bacterial expression systems

    • Solution: Reduce induction temperature (16-20°C), decrease inducer concentration, co-express with chaperones, or use detergents for membrane proteins

  • Protein degradation

    • Challenge: Proteolytic degradation during expression or purification

    • Solution: Add protease inhibitors, use protease-deficient host strains, optimize buffer conditions, and reduce purification time

  • Post-translational modification differences

    • Challenge: Heterologous systems may lack Dictyostelium-specific modifications

    • Solution: Express in eukaryotic systems or in Dictyostelium itself for critical modifications, verify modification status by mass spectrometry

  • Protein misfolding

    • Challenge: Incorrect folding leading to non-functional protein

    • Solution: Implement slow refolding protocols, use molecular chaperones, or test different buffer conditions to promote proper folding

ChallengeCommon SymptomsTroubleshooting ApproachSuccess Indicators
Low expressionFaint or absent bands on SDS-PAGETest multiple expression vectors, hosts, and conditionsVisible protein band at expected size
InsolubilityProtein in pellet after lysisLower temperature, co-express chaperonesProtein present in soluble fraction
DegradationMultiple smaller bands on gelAdd protease inhibitors, optimize purification speedSingle band at expected molecular weight
Improper foldingInactive protein, aggregationRefolding from inclusion bodies, stability screensFunctional activity in biochemical assays
Toxicity to hostPoor growth of expression cultureUse tightly regulated inducible promotersNormal growth until induction

The successful commercial production of recombinant Dictyostelium proteins demonstrates these challenges can be overcome with systematic optimization .

How should I approach contradictory data when characterizing an uncharacterized protein?

When faced with contradictory data during protein characterization, apply this methodological framework:

  • Verify experimental quality

    • Reassess all controls to ensure they performed as expected

    • Check for technical issues in experimental procedures

    • Evaluate reagent quality and specificity

    • Consider statistical power and sample size adequacy

  • Cross-validate with independent methods

    • Apply alternative techniques to address the same question

    • Use orthogonal approaches that rely on different principles

    • Compare results across different experimental systems

    • Seek independent verification from collaborators

  • Systematic analysis of variables

    • Identify factors that differ between contradictory experiments

    • Test each variable independently to isolate the source of variation

    • Consider context-dependent effects (cell type, developmental stage)

    • Evaluate dose-dependency or time-course effects

  • Consider biological complexity

    • Examine if the protein has multiple functions in different contexts

    • Investigate potential post-translational modifications

    • Assess if protein complexes change under different conditions

    • Evaluate potential feedback mechanisms or compensatory responses

  • Literature assessment and theoretical integration

    • Compare with published data on related proteins

    • Develop models that could explain seemingly contradictory results

    • Consult experts in the field for alternative interpretations

    • Design critical experiments to distinguish between competing models

This systematic approach transforms contradictory results from a frustration into an opportunity for deeper understanding of the protein's complex biology.

What statistical approaches are most appropriate for analyzing protein characterization data in Dictyostelium?

Selecting appropriate statistical methods is crucial for robust data interpretation when characterizing proteins like DDB_G0274935:

  • Preliminary data assessment

    • Test for normality using Shapiro-Wilk or Kolmogorov-Smirnov tests

    • Assess homogeneity of variance with Levene's test

    • Identify and handle outliers appropriately

    • Determine if parametric or non-parametric tests are required

  • Comparative analyses

    • For normally distributed data: t-tests (paired or unpaired) for two groups; ANOVA for multiple groups

    • For non-normally distributed data: Mann-Whitney U test for two groups; Kruskal-Wallis test for multiple groups

    • Apply appropriate post-hoc tests (Tukey, Bonferroni, Dunnett) for multiple comparisons

    • Consider repeated measures designs when appropriate

  • Correlation and regression analyses

    • Pearson correlation for linear relationships between variables

    • Spearman correlation for non-parametric correlations

    • Multiple regression to model relationships between dependent and multiple independent variables

    • Logistic regression for binary outcomes

  • Advanced statistical approaches

    • Principal component analysis for dimension reduction

    • Cluster analysis for identifying patterns in complex datasets

    • Bayesian approaches for integration of prior knowledge

    • Machine learning for predictive modeling with large datasets

  • Experimental design considerations

    • Power analysis to determine adequate sample size

    • Randomization to minimize bias

    • Blocking to control for known sources of variation

    • Factorial designs to test multiple variables simultaneously

When reporting results, include both the statistical significance (p-value) and effect size to provide a complete picture of the biological relevance of your findings. Visualization through appropriate graphs (box plots, scatter plots with error bars) enhances data interpretation and communication.

How can emerging technologies advance our understanding of uncharacterized proteins in Dictyostelium?

Several cutting-edge technologies show particular promise for elucidating the functions of uncharacterized proteins like DDB_G0274935:

  • Advanced CRISPR technologies

    • CRISPR activation/interference for precise gene expression control

    • Base editors and prime editors for specific nucleotide modifications

    • CRISPR screens for high-throughput functional analysis

    • Methodological approach: Adapt these technologies for use in Dictyostelium through vector and delivery optimization

  • Single-cell technologies

    • Single-cell RNA-seq to capture heterogeneity in gene expression

    • Single-cell proteomics for protein-level analysis

    • Live-cell imaging with advanced biosensors

    • Methodological approach: Develop Dictyostelium-specific protocols for cell isolation and analysis

  • Structural biology advances

    • Cryo-EM for high-resolution structure determination

    • AlphaFold2 and other AI-based structure prediction tools

    • Hydrogen-deuterium exchange mass spectrometry for dynamic structural information

    • Methodological approach: Express and purify proteins for structural studies, validate computational models experimentally

  • Multi-omics integration

    • Combined analysis of transcriptomics, proteomics, and metabolomics data

    • Network analysis to position proteins within cellular systems

    • Temporal multi-omics to capture dynamic processes

    • Methodological approach: Develop computational pipelines specific for Dictyostelium data integration

  • Spatial biology

    • Spatial transcriptomics to map gene expression in multicellular structures

    • Advanced microscopy methods (STORM, PALM, lattice light-sheet)

    • Correlative light and electron microscopy

    • Methodological approach: Adapt spatial mapping technologies for Dictyostelium's unique cellular contexts

These emerging technologies, when properly adapted to the unique characteristics of Dictyostelium, have the potential to revolutionize our understanding of previously uncharacterized proteins and their roles in cellular functions.

How might comparative analysis across species provide insights into uncharacterized protein function?

Comparative analysis across species offers powerful insights into protein function through these methodological approaches:

  • Evolutionary conservation analysis

    • Align sequences across diverse species to identify conserved domains

    • Quantify selection pressure on different protein regions

    • Construct phylogenetic trees to map evolutionary relationships

    • Methodological approach: Use progressive multiple sequence alignment algorithms followed by conservation scoring

  • Functional correlation across species

    • Compare phenotypes of orthologous gene mutations

    • Identify co-evolved gene clusters suggesting functional relationships

    • Examine conservation of protein-protein interaction networks

    • Methodological approach: Systematically compare phenotypic data from model organism databases

  • Comparative genomic context

    • Analyze synteny and gene neighborhood conservation

    • Examine co-occurrence patterns of genes across genomes

    • Compare regulatory elements controlling orthologous genes

    • Methodological approach: Apply computational genomic context analysis tools to multiple Dictyostelium species

  • Cross-species complementation

    • Test if orthologs from other species can rescue Dictyostelium mutants

    • Identify functionally equivalent domains through domain swapping

    • Evaluate conservation of subcellular localization

    • Methodological approach: Generate expression constructs with orthologs and test in knockout backgrounds

  • Comparative structural analysis

    • Compare predicted or solved structures across species

    • Identify conserved binding sites or catalytic residues

    • Map species-specific variations onto structural models

    • Methodological approach: Implement structure-based sequence alignments and conservation mapping

Comparative analysis across the available Dictyostelium species (D. discoideum, D. purpureum, D. fasciculatum, and P. pallidum) can provide particularly relevant insights due to their evolutionary relationships and adapted niches, while comparison with more distant species can highlight deeply conserved functions.

What strategies should be employed to transition from basic characterization to understanding physiological relevance?

Bridging the gap between molecular characterization and physiological significance requires a systematic methodological approach:

  • Context-dependent functional analysis

    • Examine protein function across developmental stages

    • Test under various stress conditions (starvation, osmotic stress, etc.)

    • Assess function in different cellular processes (chemotaxis, phagocytosis)

    • Methodological approach: Develop stage- and condition-specific assays to probe function in diverse contexts

  • Integration with organismal behavior

    • Connect molecular changes to cellular movements and morphogenesis

    • Quantify effects on single-cell and collective behaviors

    • Analyze impact on multicellular development

    • Methodological approach: Apply quantitative behavior analysis methods, such as those developed for chemotaxis

  • Systems-level analysis

    • Position the protein within global regulatory networks

    • Identify key pathway intersections and feedback mechanisms

    • Model effects of perturbations on system behavior

    • Methodological approach: Implement network analysis and mathematical modeling based on experimental data

  • Translational relevance exploration

    • Connect findings to conserved pathways in higher organisms

    • Identify potential disease relevance based on human orthologs

    • Explore environmental adaptations mediated by the protein

    • Methodological approach: Comparative analysis with mammalian systems, leveraging disease gene databases

  • Technology development for in vivo analysis

    • Generate conditional alleles for temporal control

    • Develop reporter systems for real-time monitoring

    • Create tissue-specific manipulations during multicellular phases

    • Methodological approach: Adapt advanced genetic tools for spatiotemporal precision in Dictyostelium

This comprehensive approach enables researchers to position molecular findings within broader biological contexts, transforming basic characterization into meaningful insights about physiological function and potential translational relevance.

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.