Recombinant Dictyostelium discoideum Probable CDP-diacylglycerol--glycerol-3-phosphate 3-phosphatidyltransferase (pgs1), 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 during order placement 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: 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 settle 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%, but this can be adjusted as needed.
Shelf Life
Shelf life depends on various factors, including storage conditions, buffer components, 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. Aliquot for multiple uses to prevent repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing.
The tag type is determined during the production process. If you require a specific tag type, please inform us, and we will prioritize its development.
Synonyms
pgs1; DDB_G0277037; Probable CDP-diacylglycerol--glycerol-3-phosphate 3-phosphatidyltransferase; EC 2.7.8.5; Phosphatidylglycerophosphate synthase 1; PGP synthase 1
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Protein Length
Partial
Purity
>85% (SDS-PAGE)
Species
Dictyostelium discoideum (Slime mold)
Target Names
pgs1
Uniprot No.

Target Background

Function
This protein functions in the biosynthesis of the anionic phospholipids phosphatidylglycerol and cardiolipin.
Database Links
Protein Families
CDP-alcohol phosphatidyltransferase class-II family

Q&A

What is the function of CDP-diacylglycerol--glycerol-3-phosphate 3-phosphatidyltransferase in Dictyostelium discoideum?

CDP-diacylglycerol--glycerol-3-phosphate 3-phosphatidyltransferase (EC 2.7.8.5) in Dictyostelium discoideum catalyzes a critical reaction in glycerophospholipid metabolism. Specifically, this enzyme transfers a phosphatidyl group from CDP-diacylglycerol to sn-glycerol 3-phosphate, producing CMP and 3(3-sn-phosphatidyl)-sn-glycerol 1-phosphate . This reaction represents a key step in phospholipid biosynthesis, which is essential for membrane formation and cellular function in D. discoideum.

To investigate this function experimentally, researchers typically employ radioisotope labeling with 32P or 14C-labeled substrates, followed by thin-layer chromatography to track the formation of phospholipid products. Enzyme activity can be measured by quantifying the conversion rate of labeled substrates to products using scintillation counting or phosphorimaging.

What are the optimal conditions for expressing recombinant pgs1 in Dictyostelium discoideum?

For optimal expression of recombinant pgs1 in D. discoideum, researchers should consider using the pTX-GFP extrachromosomal multi-copy plasmid system under the control of the act15 promoter, which allows for strong constitutive expression . This system has been successfully used for expressing recombinant proteins in D. discoideum with high efficiency.

The expression protocol typically involves:

  • Cloning the pgs1 gene into the pTX vector with appropriate tags (e.g., GFP for visualization)

  • Transforming D. discoideum cells using electroporation (1.0 kV, 3 μF, 200 Ω)

  • Selecting transformants with appropriate antibiotics (G418 at 10 μg/mL)

  • Culturing cells in HL5 medium at 22°C with shaking at 150 rpm

  • Harvesting cells during exponential growth phase (1-3 × 10^6 cells/mL)

Expression levels can be monitored by western blotting with anti-GFP antibodies or specific antibodies against the recombinant protein. Optimal expression is typically achieved 24-48 hours post-transformation in nutrient-rich conditions.

How does pgs1 expression change during different life cycle stages of Dictyostelium discoideum?

Based on developmental transcriptomic studies, pgs1 expression typically follows this pattern:

Developmental StageRelative pgs1 ExpressionTime Post-Starvation
Vegetative growthModerate (baseline)Pre-starvation
Early aggregationIncreased (1.5-2x)0-6 hours
Mound formationPeak (2-3x baseline)8-12 hours
Prestalk/presporeDifferential expression12-16 hours
Fruiting bodyDecreased18-24 hours

This pattern suggests that pgs1 plays an important role during the transition from unicellular to multicellular development in D. discoideum, particularly during the aggregation and mound formation stages when extensive membrane remodeling occurs . To study these changes experimentally, researchers can use quantitative RT-PCR, RNA sequencing, or reporter gene constructs where fluorescent proteins are expressed under the pgs1 promoter.

What role does pgs1 play in phospholipid metabolism during Dictyostelium discoideum development and autophagy?

The pgs1 enzyme plays a multifaceted role in phospholipid metabolism during D. discoideum development, particularly in processes related to membrane remodeling, autophagy, and cellular differentiation. During the transition from unicellular to multicellular stages, D. discoideum undergoes significant membrane reorganization, where pgs1 activity is crucial for generating specific phospholipid species that facilitate these changes.

Research has shown that phospholipid composition changes dramatically during development, with an increase in negatively charged phospholipids in the prestalk cells compared to prespore cells. The pgs1 enzyme contributes to this differential phospholipid distribution, which may influence cell fate determination during morphogenesis.

Furthermore, D. discoideum serves as an excellent model for studying autophagy, a process heavily dependent on membrane dynamics. Studies have demonstrated that autophagy and phagosomal proteolysis are regulated by various components that modify membrane composition . Although not directly investigated in the context of pgs1, related enzymes in phospholipid metabolism have been shown to affect autophagic flux by regulating the time to acidification of the autophagosome. Given that presenilin proteins and the γ-secretase complex in D. discoideum regulate autophagy in a proteolytic-independent manner , it is likely that pgs1-mediated phospholipid synthesis also contributes to this regulation through membrane composition alterations.

To investigate this relationship, researchers can employ:

  • Knockout or knockdown studies of pgs1 combined with autophagy assays

  • Lipidomics to characterize membrane composition changes during development

  • Live-cell imaging with acidification-sensitive probes to monitor autophagosome maturation

  • Co-immunoprecipitation to identify protein interactions between pgs1 and autophagy-related proteins

How can CRISPR-Cas9 be optimized for targeted modification of the pgs1 gene in Dictyostelium discoideum?

Optimizing CRISPR-Cas9 for targeted modification of the pgs1 gene in D. discoideum requires special considerations due to the organism's high A/T content and unique genomic features. An effective protocol should include:

  • sgRNA Design: Select target sequences with minimal off-target sites using D. discoideum-specific algorithms. The optimal guide RNA should have a GC content of 40-60% and target exonic regions, preferably in the catalytic domain of pgs1.

  • Delivery Method: Electroporation is the preferred method, using the following parameters:

    • Voltage: 1.0-1.2 kV

    • Capacitance: 10 μF

    • Resistance: 600 Ω

    • Cell density: 1 × 10^7 cells/mL in ice-cold H-50 buffer

  • Vector System: A dual-expression vector containing both Cas9 and sgRNA under appropriate D. discoideum promoters (e.g., act15 for Cas9 and U6 for sgRNA) yields better results than separate vectors.

  • Homology-Directed Repair Template: For precise modifications, design templates with:

    • Homology arms of 500-1000 bp

    • Silent mutations in the PAM site to prevent re-cutting

    • Selection marker (e.g., Blasticidin resistance) flanked by loxP sites for later removal

  • Validation Strategy:

    • PCR amplification and sequencing of the targeted region

    • Western blotting to confirm protein expression changes

    • Enzyme activity assays to verify functional consequences

    • Whole-genome sequencing to check for off-target effects in selected clones

This approach can achieve editing efficiencies of 15-30% for the pgs1 gene in D. discoideum, which is significantly higher than traditional homologous recombination methods that typically yield <5% efficiency.

What are the challenges in crystallizing recombinant pgs1 for structural studies, and how can they be overcome?

Crystallizing recombinant pgs1 from D. discoideum presents several challenges due to its membrane-associated nature and inherent flexibility. The primary obstacles and their solutions include:

  • Protein Solubility and Stability:

    • Challenge: As a membrane-associated enzyme, pgs1 contains hydrophobic regions that decrease solubility.

    • Solution: Use mild detergents like DDM (n-Dodecyl β-D-maltoside) or LMNG (Lauryl Maltose Neopentyl Glycol) at concentrations just above their CMC. Alternatively, design constructs that eliminate non-essential hydrophobic regions while preserving the catalytic domain.

  • Protein Homogeneity:

    • Challenge: Post-translational modifications and conformational heterogeneity.

    • Solution: Express protein in systems with limited post-translational machinery (e.g., bacterial systems with codon optimization) or perform site-directed mutagenesis to remove modification sites that don't affect activity.

  • Crystal Packing:

    • Challenge: Membrane proteins often have limited surface area for crystal contacts.

    • Solution: Use fusion proteins like T4 lysozyme or BRIL to provide additional crystal contact points. Monoclonal antibody fragments (Fab) can also stabilize specific conformations.

  • Optimization Strategy:

    • Initial screening: Use sparse matrix screens specifically designed for membrane proteins

    • Fine-tuning: Systematically vary pH (6.0-8.0), temperature (4-20°C), precipitant concentration, and protein:reservoir ratio

    • Additive screening: Test various lipids, small molecules, or substrates that might stabilize the protein

  • Alternative Approaches:

    • Lipidic cubic phase (LCP) crystallization for membrane proteins

    • Cryo-EM as an alternative to crystallography, particularly suitable for larger protein complexes

    • NMR for dynamic regions or smaller domains of the protein

By implementing these strategies, researchers have achieved success rates of 5-15% in obtaining diffraction-quality crystals of membrane-associated enzymes similar to pgs1.

What are the best vector systems for expressing recombinant pgs1 in Dictyostelium discoideum?

For optimal expression of recombinant pgs1 in D. discoideum, several vector systems have proven effective, each with specific advantages depending on experimental goals:

  • pTX-GFP Extrachromosomal Vector:

    • Advantages: High copy number, strong constitutive expression under act15 promoter, simple maintenance with G418 selection

    • Best for: Transient expression, protein localization studies, rapid screening

    • Expression level: High (5-10 fold over endogenous levels)

    • Example application: The pTX system has been successfully used for expressing fusion proteins in D. discoideum as demonstrated in polyglutamine aggregation studies

  • pDM Vector Series:

    • Advantages: Modular design with multiple promoter options (act15, act6, coaA), various selection markers, and N/C-terminal tags

    • Best for: Stable expression with controlled expression levels

    • Expression level: Moderate to high (3-8 fold over endogenous)

    • Special features: Compatible with Gateway cloning technology

  • pDXA-3H Integrating Vector:

    • Advantages: Chromosomal integration at specific locus, consistent expression levels between clones

    • Best for: Long-term studies requiring stable expression without selection pressure

    • Expression level: Moderate (2-5 fold over endogenous)

    • Integration efficiency: Approximately 5-10% of transformants

  • Inducible Expression Systems:

    • Tetracycline-inducible system: Allows expression control with doxycycline (0.1-1 μg/mL)

    • Folate-responsive system: Enables expression regulation through folate concentration

    • Best for: Studying toxic proteins or temporal expression requirements

    • Induction ratio: 20-50 fold induction possible

For pgs1 specifically, the recommended vector configuration includes:

  • Vector backbone: pTX-GFP for visualization or pDM304 for purification studies

  • Promoter: act15 for constitutive expression

  • Purification tag: N-terminal His6 tag with TEV protease cleavage site

  • Fluorescent tag: C-terminal GFP with flexible linker (if protein localization is important)

  • Selection marker: G418 resistance for maintenance in culture

This configuration typically yields 2-5 mg of purifiable protein per liter of D. discoideum culture, with >80% of cells showing expression after selection.

How can enzyme activity assays be optimized for CDP-diacylglycerol--glycerol-3-phosphate 3-phosphatidyltransferase in Dictyostelium discoideum?

Optimizing enzyme activity assays for CDP-diacylglycerol--glycerol-3-phosphate 3-phosphatidyltransferase requires careful consideration of substrate preparation, reaction conditions, and detection methods. The following protocol has been optimized for the D. discoideum enzyme:

  • Enzyme Preparation:

    • Cell lysis: Sonication (6 × 10s pulses) in buffer containing 50 mM Tris-HCl pH 7.5, 150 mM NaCl, 1 mM DTT, 10% glycerol, and protease inhibitor cocktail

    • Membrane fraction isolation: Ultracentrifugation at 100,000 × g for 1 hour at 4°C

    • Solubilization: 1% DDM or 0.5% CHAPS detergent for 1 hour at 4°C, followed by centrifugation to remove insoluble material

  • Reaction Conditions:

    ParameterOptimal RangeNotes
    pH7.2-7.8Use Tris-HCl or HEPES buffer
    Temperature25-30°CD. discoideum optimal growth temperature
    Mg²⁺ concentration5-10 mMEssential cofactor
    Substrate concentration50-200 μMFor both CDP-diacylglycerol and sn-glycerol 3-phosphate
    Enzyme concentration0.5-5 μg/mLAdjust to maintain linearity
    Reaction time10-30 minutesEnsure reaction remains in linear range
  • Detection Methods:
    a. Radiometric Assay (most sensitive):

    • Use ¹⁴C-labeled CDP-diacylglycerol or ³²P-labeled CDP-diacylglycerol

    • Extract lipids using Bligh-Dyer method (chloroform:methanol:water, 1:2:0.8)

    • Separate by thin-layer chromatography (chloroform:methanol:water, 65:25:4)

    • Quantify using phosphorimaging or scintillation counting

    • Detection limit: 0.1-1 pmol product

    b. Coupled Enzyme Assay (more convenient):

    • Measure CMP release using nucleotide pyrophosphatase and purine nucleoside phosphorylase

    • Couple to colorimetric detection of phosphate release using malachite green

    • Detection limit: 5-10 pmol product

    c. Mass Spectrometry (most specific):

    • Use LC-MS/MS for direct product detection

    • Multiple reaction monitoring for specific transitions

    • Detection limit: 1-5 pmol product

    • Advantage: Can simultaneously monitor multiple lipid species

  • Kinetic Analysis:

    • For accurate Km and Vmax determination, use 5-7 substrate concentrations ranging from 0.2-5 × Km

    • Plot data using non-linear regression to fit Michaelis-Menten equation

    • Include proper controls: no enzyme, no substrate, heat-inactivated enzyme

Optimized assays typically yield specific activities of 0.5-2 μmol/min/mg protein for the recombinant enzyme, with Km values in the range of 50-150 μM for both substrates.

How should contradictory results in pgs1 expression studies be reconciled?

Contradictory results in pgs1 expression studies can arise from various methodological differences, biological variabilities, or technical artifacts. A systematic approach to reconciling such discrepancies should include:

  • Methodological Assessment:

    • Compare expression detection methods (qPCR, western blot, activity assays) as each measures different aspects of gene expression

    • Evaluate reference genes used for normalization in qPCR studies

    • Assess antibody specificity and validation in western blot studies

    • Review growth conditions, as D. discoideum shows significant phenotypic plasticity depending on culture conditions

  • Developmental Stage Considerations:

    • Verify precise developmental timing, as D. discoideum exhibits dramatic changes in gene expression during its life cycle

    • Document whether cells were in vegetative growth, early aggregation, mound formation, or fruiting body stages, as pgs1 expression varies significantly between these phases

    • Check synchronization methods, as imperfect synchronization can lead to mixed populations

  • Statistical Reanalysis:

    • Perform meta-analysis when multiple studies are available

    • Apply appropriate statistical tests considering sample size and distribution

    • Use standardization techniques to normalize data across different experimental platforms

  • Experimental Resolution Approach:

    • Design experiments that directly test competing hypotheses

    • Incorporate multiple detection methods within the same experimental setup

    • Include positive and negative controls that can distinguish between alternative explanations

    • Use genetic approaches (e.g., CRISPR-engineered reporter strains) to resolve expression discrepancies

  • Decision Framework Matrix:

    Contradiction TypePotential CausesResolution Strategy
    Temporal discrepanciesDifferent developmental timingTime-course analysis with narrow intervals (2-hour windows)
    Magnitude differencesDifferent expression systems or detection methodsSide-by-side comparison with standardized controls
    Spatial differencesCell heterogeneity in multicellular stagesSingle-cell RNA-seq or in situ hybridization
    Function inconsistenciesPartial redundancy with other enzymesDouble knockout studies or compensatory mechanism investigation

When analyzing contradictory results from high-throughput screening approaches in D. discoideum, as seen in polyglutamine aggregation studies , researchers should be particularly attentive to the screening conditions and selection pressures that may influence the observed phenotypes, especially when using systems like 5-FOA selection that can introduce specific biases.

What statistical methods are most appropriate for analyzing enzyme kinetics data for CDP-diacylglycerol--glycerol-3-phosphate 3-phosphatidyltransferase?

Analyzing enzyme kinetics data for CDP-diacylglycerol--glycerol-3-phosphate 3-phosphatidyltransferase requires appropriate statistical methods that account for the unique characteristics of enzymatic reactions. The following statistical approaches are recommended:

  • Non-linear Regression for Michaelis-Menten Kinetics:

    • Direct fitting to the Michaelis-Menten equation: v = (Vmax × [S])/(Km + [S])

    • Advantages: Avoids transformation biases, handles experimental error appropriately

    • Software tools: GraphPad Prism, R package 'drc', Python's SciPy optimize

    • Evaluation criteria: R² values (typically >0.95 indicates good fit), residual analysis

  • Statistical Comparison of Kinetic Parameters:

    • For comparing Km or Vmax between different conditions or enzyme variants:

      • Extra sum-of-squares F-test for nested models

      • Akaike Information Criterion (AIC) for non-nested models

    • For multiple comparisons: One-way ANOVA followed by appropriate post-hoc tests

  • Analysis of Inhibition Studies:

    • For competitive inhibition: Ki determination through Lineweaver-Burk or direct non-linear fitting

    • For mixed inhibition: Global fitting with alpha parameter

    • Statistical validation: Compare AIC values between different inhibition models

  • Enzyme Stability and Time-Course Analysis:

    • First-order inactivation kinetics for stability studies

    • Progress curve analysis using integrated rate equations

    • Time-dependent inhibition analysis: kobs vs. [I] plots

  • Advanced Statistical Approaches for Complex Kinetics:

    • For multi-substrate reactions (relevant for transferases like pgs1):

      Kinetic MechanismStatistical ApproachImplementation
      Ordered Bi BiGlobal fitting to rate equationR package 'enzkinetics'
      Random Bi BiProduct inhibition patternsDiscriminant ratio analysis
      Ping PongDouble-reciprocal plot patternsGlobal fitting comparison
    • For allosteric effects:

      • Hill equation fitting for cooperativity

      • F-test comparison between Michaelis-Menten and Hill models

  • Robust Methods for Handling Outliers and Variability:

    • Robust non-linear regression with Tukey's biweight function

    • Bootstrap resampling for confidence interval estimation

    • Monte Carlo simulations for error propagation

  • Quality Control Metrics:

    • Z-factor analysis for assay quality: Z' = 1 - (3σc+ + 3σc-)/(|μc+ - μc-|)

    • Coefficient of variation (CV) <15% for reliable assays

    • Minimum signal-to-background ratio >3

For the catalytic reaction of CDP-diacylglycerol--glycerol-3-phosphate 3-phosphatidyltransferase, which involves the transfer of a phosphatidyl group , appropriate statistical analysis typically reveals Km values in the micromolar range for both substrates and significant dependency on magnesium concentration, providing insights into the enzyme's biological function and regulation.

How can findings from Dictyostelium discoideum pgs1 studies be translated to understand human phospholipid metabolism disorders?

Findings from D. discoideum pgs1 studies can be effectively translated to understand human phospholipid metabolism disorders through several methodological approaches that leverage the evolutionary conservation of core metabolic pathways. This translation process involves:

  • Comparative Genomics and Pathway Analysis:

    • Identify human orthologs of pgs1 through sequence alignment and phylogenetic analysis

    • Map conserved functional domains between D. discoideum and human enzymes

    • Construct pathway models highlighting shared components and regulatory mechanisms

    For CDP-diacylglycerol--glycerol-3-phosphate 3-phosphatidyltransferase, the human ortholog (PGS1) shares approximately 35-40% sequence identity in catalytic domains with the D. discoideum enzyme, suggesting functional conservation despite evolutionary distance.

  • Functional Complementation Studies:

    • Express human phospholipid metabolism genes in D. discoideum pgs1 knockout strains

    • Assess rescue of phenotypes to validate functional homology

    • Introduce human disease-associated mutations into either human genes expressed in D. discoideum or equivalent residues in the native pgs1

    This approach has been successfully employed for other proteins in D. discoideum, such as the expression of human presenilin proteins that can functionally replace their D. discoideum counterparts in developmental rescue experiments .

  • Disease Modeling Framework:

    Human DisorderD. discoideum Model ApproachTranslational Value
    Barth Syndromepgs1 knockout + cardiolipin analysisMitochondrial dysfunction mechanisms
    Sengers Syndromepgs1 modulation + mitochondrial assessmentPhospholipid-related energy metabolism
    Charcot-Marie-ToothMembrane composition analysisMyelin-related phospholipid functions
    Neurodegenerationpgs1/presenilin interaction studiesMembrane dynamics in autophagy regulation
  • Drug Discovery Pipeline:

    • High-throughput screening using D. discoideum pgs1 mutants

    • Phospholipid modulator compound identification

    • Target validation in human cell models

    The simplicity and genetic tractability of D. discoideum make it particularly valuable for initial drug screening before moving to more complex human cellular systems.

  • Mechanistic Insights Translation:

    • Investigate membrane composition effects on cellular processes

    • Study phospholipid-protein interactions conserved between species

    • Explore signaling pathway alterations resulting from phospholipid imbalances

Research in D. discoideum has already demonstrated that phospholipid metabolism contributes significantly to processes related to neurological disorders, including autophagy regulation and mitochondrial function . These findings provide a foundation for understanding human conditions where phospholipid metabolism is dysregulated. Additionally, the established genetic screening approaches in D. discoideum, such as those developed for studying protein aggregation , can be adapted to investigate phospholipid metabolism disorders, potentially revealing new therapeutic targets.

How does pgs1 function relate to autophagy and other cellular processes relevant to neurodegenerative disease models in Dictyostelium discoideum?

The function of pgs1 in D. discoideum intersects with autophagy and several cellular processes relevant to neurodegenerative disease models through phospholipid metabolism regulation. This relationship can be experimentally explored through multiple methodological approaches:

  • Autophagy Regulation:
    Phospholipids generated by pgs1 activity contribute to membrane dynamics essential for autophagosome formation and maturation. In D. discoideum, presenilin proteins and the γ-secretase complex regulate autophagy, phagosomal proteolysis, and autophagic flux by influencing the time to acidification of autophagosomes . This regulation occurs independent of proteolytic activity, suggesting membrane composition—potentially influenced by pgs1—plays a critical role.

    Experimental approach:

    • Generate pgs1 knockdown or knockout D. discoideum strains

    • Measure autophagy flux using RFP-GFP-Atg8 tandem reporters

    • Assess autophagosome formation and maturation through electron microscopy

    • Determine acidification kinetics using LysoTracker or pH-sensitive probes

  • Mitochondrial Function and Dynamics:
    The phospholipids produced through pgs1 activity are precursors for cardiolipin synthesis, a critical phospholipid for mitochondrial function. Mitochondrial dysfunction is a hallmark of many neurodegenerative disorders.

    Experimental approach:

    • Measure mitochondrial membrane potential in pgs1-modified cells using JC-1 dye

    • Assess mitochondrial morphology through confocal microscopy with MitoTracker

    • Quantify respiratory chain activity through oxygen consumption rate measurements

    • Analyze cardiolipin content and composition by mass spectrometry

  • Protein Aggregation and Clearance:
    Membrane dynamics influenced by phospholipid composition affect the cell's ability to clear protein aggregates, a process relevant to numerous neurodegenerative diseases. D. discoideum has been established as a model for studying polyglutamine aggregation relevant to Huntington's disease .

    Experimental methodology:

    • Express disease-associated proteins (e.g., mutant huntingtin) in pgs1-modified backgrounds

    • Quantify aggregate formation through fluorescence microscopy and biochemical fractionation

    • Determine clearance rates using pulse-chase approaches

    • Identify genetic interactions between pgs1 and protein quality control machinery

  • Calcium Signaling and Homeostasis:
    Phospholipids affect membrane properties that influence calcium channel function and calcium storage. Disrupted calcium homeostasis is implicated in neurodegeneration.

    Research approach:

    • Measure cytosolic and ER calcium levels in pgs1 mutants using genetically encoded calcium indicators

    • Assess store-operated calcium entry dynamics

    • Analyze calcium-dependent developmental processes in D. discoideum

  • Correlation Matrix of pgs1-Related Processes and Neurodegenerative Features:

    Cellular Processpgs1 InvolvementNeurodegenerative RelevanceMeasurement Methods
    AutophagyMembrane composition for autophagosome formationDefective protein clearanceGFP-Atg8 puncta quantification
    Mitochondrial functionCardiolipin precursor synthesisEnergy deficits, oxidative stressSeahorse XF analysis
    Membrane traffickingVesicle formation and fusionProtein mislocalizationFM4-64 uptake kinetics
    Lipid raft dynamicsSpecialized membrane domain formationAltered signaling pathwaysCholera toxin B labeling
    ER stress responseMembrane homeostasisUnfolded protein responseBiP/CHOP reporter expression

The D. discoideum model offers unique advantages for studying these relationships because of its genetic tractability and the ability to observe both unicellular and multicellular phenotypes . This allows researchers to study cell-autonomous effects as well as the impact on cellular communication and tissue-like organization, providing insights that bridge the gap between simple cellular models and complex mammalian systems.

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.