KEGG: ago:AGOS_AFR064C
STRING: 33169.AAS53435
Ashbya gossypii FK506-binding protein 1 (FPR1) is a gene that encodes a peptidyl-prolyl cis-trans isomerase (PPIase) belonging to the immunophilin family. The protein product (FKBP12) has a conserved structure comprising five to six β-sheets that wrap around a central α-helix, with three extended loops (40s, 50s, and 80s loops) surrounding the binding pocket. In filamentous fungi like A. gossypii, FKBP12 serves as the cellular receptor for immunosuppressive drugs such as FK506 (tacrolimus) .
The biological role of FKBP12 in A. gossypii includes protein folding, signal transduction, and possibly involvement in self-substrate recognition through dimerization. The protein can form dimers through a unique interaction where the 80s loop from one monomer docks into the active site of the second monomer, a characteristic observed in other filamentous fungi such as Aspergillus fumigatus .
A. gossypii FKBP12 shares significant structural homology with FKBP12 from other fungi and humans, but with notable differences that influence drug binding and protein-protein interactions:
| Feature | A. gossypii FKBP12 (inferred) | A. fumigatus FKBP12 | Human FKBP12 | C. albicans FKBP12 |
|---|---|---|---|---|
| Core structure | β-sheets wrapping central α-helix | β-sheets wrapping central α-helix | β-sheets wrapping central α-helix | β-sheets wrapping central α-helix |
| 40s loop | Contains fungal-specific residues | Contains F22, Q50, R55 | Contains T22, M50, E55 | Contains fungal-specific residues |
| 80s loop | Contains proline residue for dimerization | P90 facilitates dimerization | H88 instead of F88; no dimerization | P104 in cis conformation |
| FK506 binding | Binds FK506 | F88 crucial for FK506-binding | H88 crucial for FK506-binding | Similar to A. fumigatus |
| Dimerization | Likely dimerizes via 80s loop | Dimerizes via 80s loop | Does not dimerize | Dimerizes via 80s loop in cis conformation |
Based on homology with other fungal FKBP12 proteins, A. gossypii FKBP12 likely contains critical residues that distinguish it from human FKBP12, particularly in the 40s and 80s loops, which affect drug binding and protein-protein interactions .
When A. gossypii is subjected to secretion stress (induced by DTT), it experiences substantial transcriptional changes. Unlike the conventional unfolded protein response (UPR) observed in other fungi, A. gossypii displays alternative protein quality control mechanisms. This includes up-regulation of genes involved in protein unfolding, endoplasmic reticulum-associated degradation, proteolysis, vesicle trafficking, vacuolar protein sorting, and mRNA degradation .
For efficient cloning and expression of recombinant A. gossypii FPR1, researchers can follow this optimized protocol:
Cloning Strategy:
Amplify the A. gossypii FPR1 gene from genomic DNA or cDNA using high-fidelity PCR.
Design primers with appropriate restriction sites or overhangs for downstream cloning.
Consider using CRISPR/Cas9 for precise manipulation of the FPR1 gene within the A. gossypii genome itself .
Expression Systems:
For structural studies: E. coli BL21(DE3) with pET or pGEX vectors (GST-tagged or His-tagged constructs)
For functional studies: S. cerevisiae expression systems may be preferable due to phylogenetic proximity to A. gossypii
Expression Optimization:
Induce expression at lower temperatures (16-20°C) to enhance protein folding
Consider codon optimization for the expression host
Include a cleavable tag (His, GST, or MBP) to facilitate purification while allowing tag removal
Purification Protocol:
Cell lysis using sonication or French press in buffer containing protease inhibitors
Affinity chromatography using the appropriate resin for the tag
Tag cleavage if necessary
Size exclusion chromatography for final purification
While these recommendations are inferred from general recombinant protein techniques and knowledge of A. gossypii's phylogenetic relationship to yeast, specific optimization may be required for the A. gossypii FPR1 protein .
The CRISPR/Cas9 system has been adapted for A. gossypii genome editing and can be effectively used to modify the FPR1 gene:
CRISPR/Cas9 Protocol for A. gossypii FPR1 Modification:
Design of guide RNA (gRNA):
Identify target sequences within the FPR1 gene with a 5′-NGG-3′ PAM sequence
Design gRNA with 20 nucleotides complementary to the target sequence
Verify specificity using bioinformatic tools to avoid off-target effects
Construction of CRISPR/Cas9 Vector:
Use the one-vector CRISPR/Cas9 system adapted for A. gossypii
Clone the gRNA expression cassette into the vector containing Cas9
If making specific mutations, include a repair template with desired modifications
Transformation and Selection:
Transform A. gossypii spores with the CRISPR/Cas9 vector
Select transformants using appropriate markers
Verify genome modifications by PCR and sequencing
Marker-free Engineering (if desired):
The one-vector system allows for marker-free engineering strategies
Design the repair template to include desired mutations without selection markers
Verify modifications through phenotypic or molecular screening
This approach allows for precise manipulation of the FPR1 gene, including point mutations to study structure-function relationships, gene deletions to assess essentiality, or tagging for localization or purification studies .
For assessing the peptidyl-prolyl isomerase (PPIase) activity of recombinant A. gossypii FKBP12, several complementary methodologies are recommended:
Spectrophotometric Chymotrypsin-Coupled Assay:
Use synthetic tetrapeptide substrates containing proline (e.g., Suc-Ala-Leu-Pro-Phe-pNA)
Monitor cis-to-trans isomerization by measuring the release of p-nitroaniline at 390 nm
Conduct assays with and without FK506 to assess inhibition
Calculate kinetic parameters (kcat/KM) to quantify catalytic efficiency
Protease-Free NMR-Based Assay:
Use 15N-labeled peptide substrates
Monitor cis/trans isomerization by 2D 1H-15N HSQC NMR spectroscopy
Calculate isomerization rates from time-dependent spectral changes
This technique provides direct measurement without protease coupling
FK506 Binding Assays:
Isothermal titration calorimetry (ITC) to determine binding affinities (KD)
Thermal shift assays to assess protein stability upon ligand binding
Surface plasmon resonance (SPR) for real-time binding kinetics
Comparative Analysis:
Compare the activity of A. gossypii FKBP12 with that of other fungal and human FKBP12 proteins using the same substrates and conditions to identify species-specific differences in catalytic efficiency and inhibitor sensitivity.
These methodologies enable comprehensive characterization of both the enzymatic activity and drug-binding properties of A. gossypii FKBP12, providing insights into its functional properties.
Based on studies with other fungal FKBP12 proteins, mutations in the 40s, 50s, and 80s loops of A. gossypii FKBP12 would likely have significant effects on its interactions with FK506 and calcineurin:
40s Loop Mutations:
F22T, Q50M, and R55E substitutions (corresponding to A. fumigatus positions) would likely impact FK506-FKBP12 interactions with calcineurin
In A. fumigatus, mutations in the 40s loop (F37M/L) decreased calcineurin binding and increased resistance to FK506
50s Loop Mutations:
W60V mutation (based on A. fumigatus) would likely decrease calcineurin binding
This mutation in A. fumigatus increased contacts in the 80s loop, resulting in higher resistance to FK506
80s Loop Mutations:
The 80s loop is particularly critical as it contains a proline residue involved in FKBP12 dimerization
F88H mutation (corresponding to the difference between fungal and human FKBP12) would likely result in significant increase in resistance to FK506 due to reduced binding of FK506-FKBP12 complex with calcineurin
P90G (or equivalent position in A. gossypii) would likely disrupt dimerization and alter FK506 binding
These structure-function relationships highlight potential targets for developing antifungal compounds with specificity for fungal FKBP12 while minimizing interaction with human FKBP12.
The key differences in FK506 binding between A. gossypii FKBP12 (inferred from other fungal FKBP12 proteins) and human FKBP12 present opportunities for selective drug development:
Critical Structural Differences:
| Feature | Fungal FKBP12 | Human FKBP12 | Functional Consequence |
|---|---|---|---|
| 40s loop residues | F22, Q50, R55 | T22, M50, E55 | Altered FK506 binding pocket |
| 80s loop key residue | F88 | H88 | Different modes of interaction with calcineurin |
| Dimerization capability | Self-dimerizes via 80s loop | No dimerization | Unique fungal structural feature |
| 80s loop conformation | Contains proline in variable conformations | Different loop structure | Affects binding pocket accessibility |
Exploitation for Drug Development:
Design FK506 analogs that interact preferentially with F88 in fungal FKBP12 rather than H88 in human FKBP12
Develop compounds that target the unique dimerization interface of fungal FKBP12
Create modified compounds like L685,818 (a C18 hydroxy, C21 ethyl derivative of FK506) that maintain antifungal activity while reducing immunosuppressive effects
Design molecules that exploit differences in the 40s loop residues to enhance selectivity
These structural differences provide a foundation for developing non-immunosuppressive antifungal agents that selectively target fungal FKBP12 while minimizing interaction with human FKBP12 .
Dimerization of A. gossypii FKBP12, inferred from studies on other fungal FKBP12 proteins, has significant implications for its biological activity and drug interactions:
Mechanism of Dimerization:
In fungal FKBP12 proteins, the 80s loop from one monomer docks into the active site of the second monomer and vice-versa
A specific proline residue at the tip of the 80s loop (P90 in A. fumigatus or equivalent in A. gossypii) facilitates this dimerization
This dimerization appears to be unique to fungal FKBP12 and is not observed in human FKBP12
Functional Implications:
Auto-regulation: Dimerization may serve as an auto-regulatory mechanism by blocking the active site
Drug competition: The self-substrate region overlaps with the FK506 binding region, meaning FK506 must compete with dimerization
Resistance mechanism: Mutations affecting dimerization (such as P90G in A. fumigatus) confer resistance to FK506, indicating a relationship between dimerization and drug binding
Species-specific differences: In C. albicans, all FKBP12 proteins have P104 in cis conformation, while in A. fumigatus, the proline can be in either cis or trans states, suggesting species-specific dimerization dynamics
Implications for Drug Design:
Compounds that specifically disrupt fungal FKBP12 dimerization might represent a novel class of antifungals
Understanding the dynamic between dimerization and drug binding could inform the design of more potent inhibitors
The species-specific differences in dimerization dynamics could potentially be exploited for selective targeting of particular fungal pathogens
These insights highlight dimerization as a unique feature of fungal FKBP12 proteins that has significant implications for drug binding and could be exploited for selective antifungal development .
The A. gossypii FKBP12-FK506-calcineurin complex likely exhibits important differences from the human complex based on findings from other fungal species:
Structural Differences:
| Feature | Fungal Complex | Human Complex | Implication |
|---|---|---|---|
| Key binding residue | F88 in FKBP12 is critical for calcineurin binding | H88 in FKBP12 is critical for calcineurin binding | Different molecular interactions at the interface |
| Loop conformations | Different 40s and 80s loop structures | Human-specific loop structures | Altered binding pocket geometry |
| Calcineurin binding interface | Species-specific interface residues | Human-specific interface residues | Potential for selective targeting |
| Dimerization effect | Dimerization via 80s loop affects FK506 binding | No dimerization effect | Unique fungal regulatory mechanism |
Implications for Antifungal Selectivity:
FK506 Analogs:
Exploitable Differences:
The F88 vs. H88 difference presents a clear target for selective inhibitor design
Variations in the 40s loop (F22 vs. T22, Q50 vs. M50, R55 vs. E55) provide additional targets for selective binding
Compounds could be designed to take advantage of the unique dimerization properties of fungal FKBP12
Cross-Species Considerations:
These structural and functional differences provide a foundation for developing antifungal compounds that selectively target fungal FKBP12-calcineurin interactions while minimizing effects on the human pathway.
To determine if calcineurin is essential for growth in A. gossypii and compare with other fungi, several experimental approaches can be used:
Genetic Approaches:
CRISPR/Cas9 Gene Disruption:
Conditional Expression Systems:
Generate strains with calcineurin genes under control of inducible/repressible promoters
Monitor growth under repressing conditions
This approach is valuable if calcineurin is essential, as it allows controlled depletion
Pharmacological Approaches:
FK506 Sensitivity Assays:
Determine minimum inhibitory concentrations (MICs) of FK506 for A. gossypii
Compare with other fungi to assess relative sensitivity
Test growth on solid and liquid media with varying FK506 concentrations
Cyclosporin A Studies:
As an alternative calcineurin inhibitor, compare effects of cyclosporin A with FK506
Differential sensitivity may reveal aspects of pathway specificity
Functional Genomics:
Transcriptomic Analysis:
Comparative Analysis:
In many pathogenic fungi, calcineurin is not essential for viability but is crucial for virulence and stress responses
A. gossypii may differ from pathogenic fungi due to its saprophytic lifestyle
Compare findings to calcineurin dependency in related species like S. cerevisiae, which is phylogenetically close to A. gossypii
These approaches would provide comprehensive insights into the role of calcineurin in A. gossypii growth and development, informing potential antifungal strategies targeting this pathway.
Non-immunosuppressive FK506 analogs would interact with A. gossypii FKBP12 in ways that exploit the structural differences between fungal and human FKBP12-calcineurin interactions:
FK506 Analog Interactions:
Antifungal Efficacy:
| FK506 Analog | Binding to Fungal FKBP12 | Calcineurin Inhibition | Immunosuppressive Activity | Antifungal Activity |
|---|---|---|---|---|
| FK506 (parent) | High affinity | Strong inhibition | High | Strong |
| L685,818 | Preserved binding | Selective for fungal complex | Significantly reduced | Maintained against fungi |
| APX879 (inference) | Designed for fungal specificity | Selective inhibition | Minimal | Enhanced selectivity for fungi |
Factors Affecting Efficacy:
Species-Specific Differences:
Resistance Mechanisms:
Mutations in the 80s loop (e.g., P90G in A. fumigatus) confer FK506 resistance
Monitoring for such mutations in A. gossypii would be important for clinical applications
Understanding the resistance mechanism allows for the design of second-generation inhibitors
These insights provide a foundation for developing antifungal agents based on FK506 that retain activity against A. gossypii while minimizing immunosuppressive effects in humans .
Researchers face several technical challenges when expressing and purifying A. gossypii FKBP12 for structural studies:
Expression Challenges:
Codon Usage Optimization:
A. gossypii has a different codon bias than common expression hosts
Codon optimization may be necessary for efficient expression in E. coli or yeast systems
Failure to optimize can lead to truncated protein products or inclusion body formation
Protein Folding:
As a PPIase, FKBP12 has specific folding requirements
Misfolding in heterologous systems may occur, especially at high expression levels
Expression at lower temperatures (16-20°C) and slower induction rates may improve folding
Expression System Selection:
Purification Challenges:
Solubility Issues:
Protein Stability:
Small proteins like FKBP12 (~12 kDa) can be susceptible to degradation
Purification buffers may need optimization to maintain stability
Inclusion of protease inhibitors and working at lower temperatures is essential
Homogeneity Requirements for Structural Studies:
Structural methods like X-ray crystallography require highly homogeneous samples
The dynamic nature of FKBP12, with its flexible loops, may complicate crystallization
Size-exclusion chromatography as a final purification step is crucial for achieving monodisperse samples
Strategy for Success:
Design fusion constructs (e.g., His-MBP-FKBP12) to enhance solubility and facilitate purification
Express in both prokaryotic and eukaryotic systems to compare yield and quality
Optimize buffer conditions using thermal shift assays to enhance stability
Consider including FK506 or analogs during purification to stabilize the protein
Employ multi-step purification including affinity, ion exchange, and size exclusion chromatography
These approaches address the specific challenges of working with A. gossypii FKBP12 and increase the likelihood of obtaining sufficient quantities of high-quality protein for structural studies.
In the absence of crystal structures, AlphaFold and other computational approaches offer powerful alternatives for predicting A. gossypii FKBP12-FK506-calcineurin interactions:
AlphaFold2 Implementation Strategy:
Individual Protein Structure Prediction:
Protein-Protein Complex Prediction:
Use AlphaFold-Multimer or RoseTTAFold-Complex to model the FKBP12-calcineurin interaction
Incorporate evolutionary constraints from multiple sequence alignments of fungal proteins
Compare predicted interfaces with known fungal FKBP12-calcineurin complexes
Ligand Docking Integration:
Employ molecular docking software (Autodock Vina, GOLD, Glide) to model FK506 binding
Use hybrid approaches combining AlphaFold predictions with traditional docking
Validate docking poses against known FK506-FKBP12 structures
Complementary Computational Approaches:
Molecular Dynamics Simulations:
Perform long-timescale simulations of predicted complexes to assess stability
Analyze binding free energies using methods like MM-PBSA or FEP
Investigate dynamics of key loops (40s, 50s, 80s) that influence binding
Machine Learning-Based Interface Prediction:
Train machine learning models on known FKBP12-calcineurin interfaces
Apply models to predict A. gossypii-specific interaction features
Integrate predictions with AlphaFold structural models
Experimental Validation Plan:
Applications of Predicted Models:
Virtual screening for A. gossypii-specific inhibitors targeting the unique aspects of its FKBP12-calcineurin interface
Rational design of mutations to test structural hypotheses about species-specific interactions
Guiding experimental structure determination by providing molecular replacement models for X-ray crystallography
This integrative computational approach offers valuable structural insights while experimental structures are being pursued, accelerating the development of A. gossypii-specific inhibitors.
Research on A. gossypii FKBP12 has several promising applications for developing novel antifungal strategies:
Selective Antifungal Development:
Non-Immunosuppressive FK506 Derivatives:
Dimerization Disruptors:
Target the unique dimerization interface observed in fungal FKBP12 proteins
Design small molecules that specifically bind to the 80s loop
Since dimerization is not observed in human FKBP12, this approach offers inherent selectivity
Fungal-Specific Calcineurin Binding:
Develop compounds that specifically interfere with the fungal FKBP12-calcineurin interface
Focus on interface residues that differ between fungal and human complexes
Biotechnological Applications:
Engineered Protein Production:
Conditional Expression Systems:
Develop FK506-responsive gene expression systems for biotechnological applications
Use engineered FKBP12 variants as molecular switches for controlling gene expression
Such systems could be valuable for controlled production of industrial enzymes
Cross-Species Applications:
Broad-Spectrum Antifungals:
Species-Selective Targeting:
Combination Therapies:
Design strategies that combine FKBP12 inhibitors with other antifungal agents
Target multiple cellular pathways simultaneously to reduce resistance development
Exploit synergistic effects between calcineurin inhibition and other antifungal mechanisms
These applications demonstrate the potential of A. gossypii FKBP12 research to contribute to both antifungal drug development and biotechnological innovation, addressing important clinical and industrial challenges.