Recombinant Neurospora crassa Ubiquitin-like protein ATG12 (atg-12)

Shipped with Ice Packs
In Stock

Description

Conjugation and E3 Ligase Activity

  1. ATG12-ATG5 Conjugation:

    • ATG12 is activated by ATG7 (E1-like enzyme), transferred to ATG10 (E2-like enzyme), and covalently linked to ATG5 .

    • The ATG12-ATG5 conjugate forms a complex with ATG16, localizing to autophagosome precursor membranes .

  2. LC3 Lipidation:

    • The ATG12-ATG5-ATG16 complex stimulates the transfer of LC3 from ATG3 to phosphatidylethanolamine (PE), enabling membrane expansion .

    • In S. cerevisiae, membrane binding is mediated by ATG5 and enhanced by ATG16 .

ATG3 Recruitment

ATG12 binds a flexible region in ATG3 via a hydrophobic pocket, recruiting the E2 enzyme to lipidation sites . Mutations in ATG3 residues (e.g., L, V, or F) disrupt this interaction and impair autophagy .

Recombinant ATG12 Production

Recombinant ATG12 is typically expressed in heterologous systems (e.g., yeast or bacterial) with affinity tags (e.g., His tag) for purification . For N. crassa, the process would involve:

  1. Cloning: Isolation of the atg-12 gene and insertion into an expression vector.

  2. Expression: Production in a suitable host (e.g., Pichia pastoris or E. coli).

  3. Purification: Affinity chromatography (e.g., Ni-NTA for His-tagged proteins) .

Table 2: Recombinant ATG12 Production Parameters

ParameterS. cerevisiae H. sapiens N. crassa (Hypothetical)
Expression HostYeastBacterial systemsP. pastoris or E. coli
Purification TagHis tag His tag His tag
YieldHighModerateVariable (dependent on host)
Purity>90% >90% Estimated >90%

Key Discoveries

  • ATG3 Binding: Structural studies of human ATG12 revealed a hydrophobic pocket that binds a β-strand/α-helix motif in ATG3 .

  • Membrane Localization: The ATG12-ATG5-ATG16 complex binds liposomes via ATG5, with ATG16 enhancing stability .

  • Proteasomal Degradation: Free ATG12 (non-conjugated) is ubiquitinated and degraded by the proteasome, linking autophagy to cell death .

Unresolved Questions for N. crassa

  1. N-terminal Function: Does the N-terminal extension of N. crassa ATG12 influence membrane localization or scaffold assembly?

  2. Species-Specific Interactions: Are ATG12-ATG3 binding residues conserved in N. crassa?

  3. Regulation of Free ATG12: Is proteasomal degradation of unbounded ATG12 conserved in N. crassa?

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 purchase 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 default glycerol concentration is 50% and can serve as a guideline.
Shelf Life
Shelf life depends on 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. 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 for preferential development.
Synonyms
atg12; NCU10049Ubiquitin-like protein atg12; Autophagy-related protein 12
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-157
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Neurospora crassa (strain ATCC 24698 / 74-OR23-1A / CBS 708.71 / DSM 1257 / FGSC 987)
Target Names
Target Protein Sequence
MASSQPLHGT ASPSLVHDDN NPNSSTASPV LEGRDSPNLP LTMTASTVLM TLPRDATAAL AEAGTFDQEK VVIRFKPVGS APALRREQVK VLSTHSFETV VAYLRKTLKV QETDSVFLYV NSVFAPALDE VVGNLWRCFK DSTNQLNVSY SMTPSFG
Uniprot No.

Target Background

Function
Recombinant *Neurospora crassa* Ubiquitin-like protein ATG12 (atg-12) is a ubiquitin-like protein involved in cytoplasm-to-vacuole transport (CVT), autophagy vesicle formation, mitophagy, and nucleophagy. Conjugation with Atg4/Atg5, via a ubiquitin-like conjugating system also involving Atg7 (E1-like activating enzyme) and Atg10 (E2-like conjugating enzyme), is crucial for its function. The Atg12-Atg4/Atg5 conjugate acts as an E3-like enzyme, essential for the lipidation of Atg8 and its association with vesicle membranes.
Database Links

KEGG: ncr:NCU10049

Protein Families
ATG12 family
Subcellular Location
Preautophagosomal structure membrane; Peripheral membrane protein.

Q&A

What is ATG12 and what role does it play in Neurospora crassa?

ATG12 is a ubiquitin-like protein that plays a critical role in the autophagy pathway, a conserved degradative process in eukaryotic organisms. In N. crassa, as in other eukaryotes, ATG12 is essential for autophagosome formation. Unlike ubiquitin, which is conjugated to multiple targets in an inducible and reversible manner, ATG12 appears to have a single target protein, ATG5, and this conjugation occurs constitutively . The ATG12-ATG5 conjugate facilitates the lipidation of another ubiquitin-like protein, ATG8, and directs its correct subcellular localization, which is crucial for membrane expansion during autophagosome formation .

How does the ATG12 conjugation system work in filamentous fungi?

The ATG12 conjugation system in filamentous fungi like N. crassa closely resembles the canonical ubiquitination pathway but with important distinctions. ATG12 is activated by an E1-like enzyme, ATG7, forming a thioester bond between the C-terminal glycine of ATG12 and a cysteine residue on ATG7 . ATG12 is then transferred to an E2-like enzyme, ATG10, and finally conjugated to a lysine residue on ATG5 through an isopeptide bond . Unlike the ubiquitin system, no typical E3 enzyme is involved in ATG12-ATG5 conjugation. Additionally, the amino acid sequence of ATG12 naturally ends with a glycine residue, eliminating the need for protease processing prior to activation . The ATG12-ATG5 conjugate further interacts with ATG16 to form a multimeric complex estimated to be approximately 350 kDa in size, representing a tetramer of ATG12-ATG5-ATG16 .

What expression systems are most suitable for producing recombinant N. crassa ATG12?

Several expression systems have been used successfully for producing recombinant ATG12 proteins, including those from N. crassa. Based on available research, the following systems have demonstrated effectiveness:

  • E. coli expression system - Suitable for producing truncated or full-length ATG12 with His-tags, achieving purities of approximately 95% .

  • Yeast expression system - Effective for expressing full-length ATG12 (AA 1-141) with His-tags, achieving purities greater than 90% .

  • Mammalian cell systems (HEK-293) - Provides higher likelihood of proper folding and post-translational modifications, with purities exceeding 90% as determined by Bis-Tris PAGE, ELISA, Western Blot, and analytical SEC .

  • Cell-free protein synthesis (CFPS) - Offers rapid production with reasonable yields (70-80% purity) and is suitable for both full-length and truncated versions of ATG12 .

For N. crassa-specific proteins, homologous expression using N. crassa itself may provide advantages for proper folding and modifications, particularly when using inducible promoter systems such as the copper-responsive tcu-1 promoter .

How can I optimize the expression and purification of functional N. crassa ATG12 for structural studies?

Obtaining high-quality, functional N. crassa ATG12 for structural studies requires careful optimization of expression and purification protocols. A methodological approach includes:

  • Expression system selection: For structural studies, E. coli systems may be sufficient for the ATG12 core domain, but eukaryotic expression systems often yield protein with more native-like folding. Consider using Saccharomyces cerevisiae or Pichia pastoris for expression, as these systems have been successfully used for other fungal ATG proteins .

  • Construct design:

    • Include the complete ubiquitin-fold region (based on crystallographic data of related ATG12 proteins)

    • For crystallization, remove flexible regions that may impede crystal formation

    • Consider fusion tags that enhance solubility (MBP, SUMO) with precision proteases for tag removal

  • Optimization steps:

    • Test multiple induction conditions (temperature, inducer concentration)

    • Screen buffer compositions during purification to maintain protein stability

    • Employ size exclusion chromatography as a final purification step to ensure monodispersity

    • Verify protein folding using circular dichroism before crystallization attempts

  • Functional verification: Assess conjugation activity with recombinant ATG5 in vitro using purified ATG7 and ATG10, which serves as both a functional test and a method to obtain the physiologically relevant ATG12-ATG5 conjugate for structural studies .

For cryo-EM studies, focus on obtaining the complete ATG12-ATG5-ATG16 complex, as this represents the biologically active form and provides insights into the structural basis of its function in autophagosome formation.

What are the distinct features of N. crassa ATG12 compared to ATG12 proteins from other model organisms?

N. crassa ATG12 shares the conserved ubiquitin-fold structural domain with ATG12 proteins from other organisms, but also exhibits distinct features reflecting its evolutionary adaptation in filamentous fungi:

Comparative structural and functional analyses between N. crassa ATG12 and its homologs provide valuable insights into both conserved mechanisms and evolutionary adaptations of autophagy across eukaryotic lineages.

How can I design a system to study ATG12 function in N. crassa using genetic manipulation?

Establishing a robust system to study ATG12 function in N. crassa requires strategic genetic manipulation approaches:

  • Gene replacement/knockout strategy:

    • Utilize homologous recombination to replace the native ATG12 gene with a marked version

    • The csr-1 locus provides an effective target for gene replacement using cyclosporin A resistance as a selectable marker

    • Design a linear construct with homology to the flanking regions of ATG12, incorporating your modification or deletion

  • Expression control systems:

    • Implement the copper-responsive tcu-1 promoter system to achieve tunable expression of ATG12

    • In copper-sufficient conditions, tcu-1 expression is repressed, while copper chelation activates expression with up to 40-fold dynamic range

    • This allows for temporal control of ATG12 expression by simply manipulating copper availability in the medium

  • Functional tagging:

    • Generate C-terminal tags (GFP, mCherry) to track localization without disrupting the critical C-terminal glycine required for conjugation

    • Use N-terminal tags with caution as they may interfere with proper processing

    • For studying protein interactions, epitope tags (FLAG, HA) can be incorporated, similar to techniques used for other N. crassa proteins

  • Verification methodology:

    • Confirm successful integration using PCR with primers spanning the integration junctions

    • Verify expression levels using quantitative RT-PCR and Western blotting

    • Assess phenotypic consequences through microscopic examination of autophagy (using additional markers like ATG8-GFP)

    • For competition experiments, implement the PCR-based high-resolution melting assay to quantify relative abundance of marked strains

  • Strain background selection:

    • The well-characterized laboratory strains FGSC 2489 and 4200 provide reliable genetic backgrounds

    • For specialized studies, consider the bd (band) background, which enhances the expression of circadian rhythms and can be useful if studying autophagy in relation to circadian control

By combining these approaches, researchers can create sophisticated genetic systems to dissect ATG12 function, including conditional expression, structure-function analyses, and interaction studies in the native N. crassa context.

What controls should be included when studying ATG12 conjugation in N. crassa?

Designing rigorous controls is essential for studying ATG12 conjugation in N. crassa:

Essential controls for ATG12 conjugation studies:

  • Genetic controls:

    • Wild-type strain (positive control for normal conjugation)

    • ATG12 deletion strain (negative control for conjugation)

    • ATG7 or ATG10 deletion strains (to confirm E1/E2 dependency)

    • ATG12 G140A mutant (replacing the C-terminal glycine with alanine to prevent conjugation)

  • Biochemical controls:

    • Reducing vs. non-reducing conditions in protein gels (the thioester bond between ATG12 and ATG7 is sensitive to reducing agents)

    • Immunoprecipitation controls using unrelated antibodies

    • Size controls to distinguish free ATG12 (~16 kDa) from ATG12-ATG5 conjugate (~70 kDa)

  • Stress induction controls:

    • Nitrogen starvation (positive control for autophagy induction)

    • Rapamycin treatment (TOR inhibitor, positive control for autophagy induction)

    • Normal growth conditions (baseline control)

  • Microscopy controls:

    • Include colocalizations with known autophagosomal markers (e.g., ATG8-tagged protein)

    • Use physiological inhibitors of autophagy as negative controls

    • Include subcellular fractionation to confirm membrane association patterns

When conducting genetic manipulations of the ATG12 system, integrate your constructs at defined genetic loci like his-3 or csr-1 to ensure consistent expression across strains . For luciferase reporter assays, include control constructs with standard AUG start codons to establish baseline expression efficiency, as has been done in N. crassa translation studies .

How can I assess the impact of environmental conditions on ATG12-mediated autophagy in N. crassa?

Evaluating environmental influences on ATG12-mediated autophagy requires methodical approaches:

  • Nutrient limitation experiments:

    • Establish a gradient of nitrogen limitation (0-100% normal levels)

    • Test carbon source depletion independently and in combination with nitrogen limitation

    • Monitor ATG12-ATG5 conjugation levels by Western blotting under each condition

    • Correlate conjugation levels with autophagosome formation using fluorescence microscopy

  • Stress response analysis:

    • Expose cultures to oxidative stress (H₂O₂), osmotic stress (NaCl), ER stress (tunicamycin)

    • Implement temperature shifts to assess heat and cold stress responses

    • Measure both acute (0-6 hours) and adaptive (24-72 hours) autophagy responses

    • Compare wild-type and ATG12-deficient strains for stress survival rates

  • Temporal regulation assessment:

    • Synchronize cultures and sample at defined intervals across the circadian cycle

    • Use the band (bd) genetic background to enhance circadian phenotypes

    • Apply the tcu-1 promoter system to control ATG12 expression at specific time points

    • Correlate autophagy activity with other circadian-regulated processes

  • Developmental stage analysis:

    • Examine ATG12 expression and conjugation across key developmental transitions:

      • Germination

      • Hyphal extension

      • Conidiation (asexual sporulation)

      • Sexual reproduction

    • Use competition experiments with marked strains to quantify fitness effects of ATG12 manipulation at each developmental stage

  • Quantification methods:

    • Implement the high-resolution melting assay for competitive fitness assessment

    • Use fluorescent ATG8 protein to quantify autophagosome numbers and size

    • Apply quantitative proteomics to measure selective cargo degradation rates

    • Employ transmission electron microscopy for ultrastructural confirmation of autophagosome formation

This systematic approach will reveal how ATG12-mediated autophagy responds to environmental cues and contributes to N. crassa's remarkable adaptability across diverse ecological niches.

What are the most effective methods for detecting ATG12-ATG5 conjugation in N. crassa?

Detecting ATG12-ATG5 conjugation in N. crassa requires specialized techniques adapted to fungal systems:

Table 1: Comparison of ATG12-ATG5 Conjugation Detection Methods in N. crassa

MethodSensitivitySpecificityQuantificationLive Cell CapabilityTechnical Difficulty
Western blottingHighHighModerateNoModerate
Mass spectrometryVery highVery highHighNoHigh
Fluorescence microscopyModerateModerate-HighModerateYesModerate
Proximity ligation assayHighHighModerateNoHigh
FRET/BRETModerateHighModerateYesHigh

Detailed methodological approaches:

  • Western blotting protocol optimization:

    • Harvest mycelia rapidly and flash-freeze in liquid nitrogen

    • Use a fungal-optimized protein extraction buffer containing protease inhibitors

    • Prepare samples in non-reducing conditions when examining thioester intermediates

    • Use gradient gels (4-12%) to resolve both free ATG12 (~16 kDa) and the conjugate (~70 kDa)

    • Detect with antibodies against N. crassa ATG12 or epitope tags if using tagged constructs

  • Epitope tagging strategy:

    • Introduce tags at positions that don't interfere with conjugation

    • For ATG12, avoid C-terminal tags that would block the critical glycine residue

    • For ATG5, avoid tagging near the Lys149 conjugation site

    • Use small epitope tags (HA, FLAG) rather than larger fluorescent proteins for biochemical assays

  • Mass spectrometry approach:

    • Immunoprecipitate ATG5 or ATG12 from N. crassa lysates

    • Perform in-gel digestion followed by LC-MS/MS analysis

    • Look for specific peptides spanning the conjugation junction

    • Use AQUA peptides as internal standards for absolute quantification

  • Fluorescence imaging optimization:

    • Create dual-labeled strains (e.g., ATG12-GFP and ATG5-mCherry)

    • Use confocal microscopy with appropriate controls for autofluorescence

    • Apply deconvolution techniques to enhance resolution

    • Quantify colocalization using Pearson's or Mander's coefficients

By combining multiple detection methods, researchers can achieve comprehensive characterization of ATG12-ATG5 conjugation dynamics in N. crassa under various experimental conditions.

What are common challenges in expressing recombinant N. crassa ATG12 and how can they be addressed?

Researchers frequently encounter several challenges when expressing recombinant N. crassa ATG12. Here are the most common issues and effective solutions:

  • Low expression levels:

    • Problem: ATG12 expression may be poor in heterologous systems.

    • Solutions:

      • Optimize codon usage for the expression host

      • Test multiple expression strains (BL21(DE3), Rosetta for E. coli; different yeast strains)

      • Implement the tcu-1 promoter system with copper chelation to boost expression in N. crassa

      • Consider cell-free protein synthesis for difficult constructs

  • Protein insolubility:

    • Problem: Recombinant ATG12 may form inclusion bodies.

    • Solutions:

      • Reduce induction temperature (16-20°C)

      • Express as fusion with solubility-enhancing tags (MBP, SUMO, TRX)

      • Test various lysis buffers with different detergents and salt concentrations

      • Consider on-column refolding protocols if necessary

  • Degradation during purification:

    • Problem: ATG12 may be susceptible to proteolysis.

    • Solutions:

      • Include protease inhibitor cocktails optimized for fungal proteases

      • Maintain samples at 4°C throughout purification

      • Add reducing agents to prevent oxidation-induced aggregation

      • Consider rapid purification methods like IMAC combined with size exclusion

  • Non-specific conjugation:

    • Problem: Recombinant ATG12 may form non-physiological conjugates.

    • Solutions:

      • Mutate the C-terminal glycine for structural studies not requiring conjugation

      • Co-express with cognate ATG5, ATG7, and ATG10 for proper conjugation

      • Verify conjugate identity by mass spectrometry

  • Inconsistent activity:

    • Problem: Batch-to-batch variation in functional activity.

    • Solutions:

      • Develop standardized activity assays based on ATG5 conjugation efficiency

      • Include internal controls in each preparation

      • Store protein in small aliquots with stabilizing agents

      • Validate each batch with multiple functional assays

For N. crassa-specific expression, leveraging the homologous recombination system with the csr-1 locus provides an effective approach for stable integration and expression . Additionally, the luciferase reporter system developed for N. crassa can be adapted to optimize and quantify expression efficiency of recombinant proteins .

How can contradictory data about ATG12 function in N. crassa be reconciled with findings from other organisms?

When faced with contradictory data about ATG12 function between N. crassa and other model organisms, a systematic approach to reconciliation is necessary:

  • Methodological differences assessment:

    • Carefully examine experimental conditions (growth media, temperature, developmental stage)

    • Consider strain background differences (laboratory vs. wild isolates)

    • Evaluate detection methods sensitivity and specificity

    • Replicate key experiments using standardized protocols across organisms

  • Evolutionary context analysis:

    • Conduct phylogenetic analyses of ATG12 and interacting partners

    • Identify lineage-specific adaptations in autophagy machinery

    • Consider the unique ecology and lifestyle of filamentous fungi versus other model organisms

    • Map functional differences to structural variations when possible

  • Functional redundancy investigation:

    • Search for potential paralogs or functionally redundant proteins in N. crassa

    • Perform double or triple knockout experiments to reveal masked phenotypes

    • Use systems biology approaches to map the complete autophagy network

    • Apply synthetic genetic array methodology to identify genetic interactions

  • Developmental timing considerations:

    • Test whether apparent contradictions result from developmental stage differences

    • Compare autophagy regulation across the complete N. crassa life cycle

    • Use the copper-inducible tcu-1 system to express ATG12 at specific developmental timepoints

    • Apply competition assays to measure fitness effects across different growth phases

  • Harmonization framework:

    • Develop integrated models that account for organism-specific variations

    • Design experiments specifically targeting contradictory findings

    • Consider that differences may reflect genuine biological adaptations rather than experimental artifacts

    • Organize community efforts to standardize research approaches

When evaluating contradictory findings, it's important to remember that N. crassa's filamentous growth pattern, robust cell wall, and unique ecological niche may necessitate specialized adaptations of conserved autophagy machinery, including ATG12-mediated processes.

What statistical approaches are most appropriate for analyzing competition experiments involving ATG12-modified N. crassa strains?

  • Bayesian approach for relative fitness estimation:

    • Implement Bayesian models that account for uncertainty in measured strain proportions

    • This approach is particularly suitable for competition experiments using marked strains

    • Incorporate prior knowledge about growth rates when available

    • Generate posterior probability distributions of fitness differences between strains

    • Calculate Bayes factors to quantify evidence for fitness effects

  • Time-series analysis methods:

    • Apply generalized linear mixed models (GLMMs) to account for temporal correlation

    • Use logistic growth curve fitting to extract key parameters (carrying capacity, exponential growth rate)

    • Implement functional data analysis for continuous monitoring data

    • Calculate selection coefficients across multiple timepoints to capture dynamics

  • Frequency-dependent selection analysis:

    • Test for frequency dependence by initiating competitions at different starting ratios

    • Apply game theory models to quantify frequency-dependent selection

    • Analyze invasion fitness by measuring growth of rare variants

    • Plot fitness as a function of frequency to identify equilibrium points

  • Environmental variation considerations:

    • Implement factorial experimental designs varying both genetic background and environment

    • Use ANOVA with interaction terms to identify GxE effects

    • Apply principal component analysis to identify major environmental response patterns

    • Calculate reaction norms to visualize fitness across environmental gradients

  • Recommended workflow for analyzing high-resolution melting assay data:

    • Normalize melting curves against internal controls

    • Apply mixture model deconvolution to estimate strain proportions

    • Calculate selection coefficients using the formula: s = (1/t)ln(pt(1-p0)/(p0(1-pt)))

    • Use bootstrapping to generate confidence intervals

    • Compare nested models with likelihood ratio tests to determine significance

Table 2: Statistical Methods Comparison for N. crassa Competition Experiments

Statistical MethodStrengthsLimitationsSuitable Experimental Design
Bayesian inferenceAccounts for uncertainty, incorporates prior knowledgeComputationally intensive, requires prior specificationHigh-resolution melting assay with multiple timepoints
Frequentist regressionSimpler implementation, widely acceptedMay not capture complex dynamics, assumes normalityDirect plating/counting with large sample sizes
Nonparametric testsNo distributional assumptions, robust to outliersLower statistical power, limited modeling optionsSmall-scale preliminary experiments
Maximum likelihoodFlexible modeling framework, handles missing dataMay converge to local optima, sensitive to initial valuesPCR-based frequency measurements with known error structure

What emerging technologies could advance our understanding of ATG12 function in N. crassa?

Several cutting-edge technologies show promise for revealing new insights into ATG12 function in N. crassa:

  • CRISPR-Cas9 genome editing:

    • Implement precise genome editing to create point mutations in key ATG12 residues

    • Generate domain swaps between N. crassa ATG12 and homologs from other species

    • Create libraries of ATG12 variants for high-throughput functional screens

    • Develop conditional knockout systems using inducible Cas9 expression

  • Proximity-dependent labeling proteomics:

    • Adapt BioID or TurboID systems for N. crassa to identify proximal proteins

    • Fuse promiscuous biotin ligases to ATG12 or ATG5 to map the autophagosome assembly interactome

    • Compare interaction networks under different nutritional or stress conditions

    • Identify novel, fungal-specific interaction partners

  • Super-resolution microscopy:

    • Apply techniques like PALM, STORM, or STED to visualize autophagosome assembly with nanometer precision

    • Implement lattice light-sheet microscopy for long-term imaging with minimal photodamage

    • Use correlative light and electron microscopy (CLEM) to connect molecular details with ultrastructural features

    • Develop N. crassa-specific fluorescent probes optimized for fungal cell imaging

  • Single-cell approaches:

    • Implement microfluidic systems for monitoring individual hyphae over time

    • Apply single-cell RNA-seq to capture cell-to-cell variation in autophagy responses

    • Develop biosensors for real-time monitoring of ATG12 activity in living cells

    • Use laser microdissection to isolate specific cellular regions for analysis

  • Synthetic biology tools:

    • Design orthogonal autophagy systems with engineered specificity

    • Create optogenetic or chemogenetic tools to control ATG12 activity with spatial and temporal precision

    • Develop synthetic genetic circuits to probe autophagy regulation

    • Engineer minimal synthetic autophagosomes to define essential components

These emerging technologies, when adapted to the unique biological context of N. crassa, will enable unprecedented insights into the function and regulation of ATG12 and its role in fungal autophagy.

How might studies of N. crassa ATG12 contribute to our understanding of fungal pathogenicity and antifungal development?

Research on N. crassa ATG12 has significant implications for understanding fungal pathogenicity and developing novel antifungal strategies:

  • Comparative autophagy analysis in pathogenic fungi:

    • N. crassa serves as a non-pathogenic model for comparing autophagy mechanisms with pathogenic filamentous fungi

    • Differences in ATG12 regulation or function between pathogenic and non-pathogenic fungi may reveal virulence-associated adaptations

    • Conservation analysis can identify fungal-specific features of ATG12 that could serve as selective targets

  • Stress response and virulence connections:

    • Autophagy mediates survival under host-imposed stresses (oxidative stress, nutrient limitation)

    • ATG12-dependent processes likely contribute to fungal persistence during host colonization

    • Competition experiments with wild-type and ATG12-modified strains under host-mimicking conditions can predict virulence factors

  • Drug target potential assessment:

    • The unique features of fungal ATG12 conjugation machinery may provide selective targets

    • High-throughput screening systems can be developed using N. crassa as a safe model

    • Structure-based drug design targeting the ATG12-ATG5 interface or ATG7-ATG12 interaction

    • Evaluation of autophagy inhibitors in combination with established antifungals for synergistic effects

  • Host-pathogen interaction insights:

    • ATG12-mediated selective autophagy may target host defense components

    • Understanding how pathogens modify autophagy pathways to evade host immunity

    • Potential application of the copper-responsive tcu-1 promoter system to model gene expression changes during infection

  • Biotechnological applications:

    • Engineering enhanced ATG12 systems for improved heterologous protein production

    • Developing fungal strains with modified autophagy for industrial applications

    • Creating biosensors based on ATG12 conjugation to detect environmental stresses or antifungal compounds

By leveraging the genetic tractability and non-pathogenic nature of N. crassa, researchers can probe fundamental aspects of ATG12 function that may translate to clinically relevant insights for combating pathogenic fungi.

What are the key considerations for researchers new to working with recombinant N. crassa ATG12?

Researchers entering the field of N. crassa ATG12 research should consider several critical factors to ensure successful investigations:

  • Experimental system selection: Carefully evaluate expression systems based on your research goals. While E. coli systems offer simplicity and high yields, eukaryotic systems may provide more physiologically relevant post-translational modifications. For structural studies, consider cell-free systems that can produce protein with 70-80% purity .

  • Genetic manipulation strategy: Leverage the homologous recombination capabilities of N. crassa using established loci like csr-1 for gene integration . The copper-responsive tcu-1 promoter system offers excellent control over gene expression with up to 40-fold dynamic range .

  • Functional validation approaches: Implement multiple complementary techniques to verify ATG12 function, including biochemical assays for conjugation, microscopy for localization, and competition experiments for fitness effects. The high-resolution melting assay provides a sensitive method for quantifying strain proportions in competition experiments .

  • Strain background considerations: Use well-characterized laboratory strains like FGSC 2489 as genetic backgrounds for consistency and reproducibility . When studying mating-type-dependent effects, note that mat a appears to have higher competitive fitness than mat A .

  • Data interpretation framework: Apply appropriate statistical methods, particularly Bayesian approaches for competition experiments, to account for uncertainty in strain proportion measurements . Consider evolutionary context when comparing results across fungal species.

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.