Recombinant Candida glabrata Histone-lysine N-methyltransferase, H3 lysine-4 specific (SET1), partial

Shipped with Ice Packs
In Stock

Description

Mechanism of Action in Azole Resistance

SET1 regulates azole (e.g., fluconazole) susceptibility by modulating ergosterol biosynthesis pathways.

Species-Specific Mechanisms

OrganismMechanism of Azole Susceptibility in SET1-Deficient StrainsClinical Relevance
C. glabrataLate ERG Pathway Downregulation: Loss of SET1 reduces expression of ERG11, ERG3, and 10 other ergosterol-related genes . H3K4me3 enrichment at ERG promoters is absent .Clinical isolates lacking SET1 show hypersusceptibility .
S. cerevisiaeEfflux Pump Inhibition: SET1 deletion reduces Pdr5 efflux pump expression .Not observed in C. glabrata .

Gene Expression and Ergosterol Metabolism

  • RNA-Seq Data: C. glabrata SET1 mutants fail to upregulate all 12 late ERG genes under azole stress .

  • Ergosterol Levels: HPLC analysis confirms SET1 is essential for maintaining ergosterol homeostasis during azole treatment .

  • ChIP-Seq: H3K4me3 is enriched at ERG gene promoters in response to azoles; this enrichment is SET1-dependent .

Catalytic Activity and Subunit Dependency

Subunit DeletionEffect on H3K4 Methylation in C. glabrataSource
SET1Complete loss of H3K4me1, me2, me3 .
BRE2Partial loss of H3K4me1/me2; no me3 .
SPP1Total loss of H3K4me3; reduced me1/me2 .

Azole Susceptibility in Clinical Isolates

  • Hypersusceptibility: C. glabrata clinical isolates lacking SET1 show reduced ERG11 expression and increased azole sensitivity .

  • Synergistic Potential: HDAC inhibitors (e.g., MGCD290) enhance azole efficacy by disrupting ERG pathways .

Virulence and Pathogenesis

  • Ergosterol Deficiency: SET1 deletion reduces ergosterol, impairing membrane integrity and adhesion .

  • Translational Targets: SET1 regulates cell wall genes; its loss may reduce virulence .

Future Directions and Research Gaps

  1. Epigenetic Therapies: Exploring SET1/ERG pathway inhibitors as adjuvants to azole drugs .

  2. Structural Studies: Mapping interactions between SET1 subunits (e.g., Spp1, Swd1) in C. glabrata .

  3. Cross-Species Comparisons: Elucidating why C. glabrata relies on SET1 for ERG regulation, unlike S. cerevisiae .

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 purchase method and location. Please contact your local distributor for precise delivery estimates.
Note: All proteins are shipped with standard 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 consolidate 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% and may serve as a guideline.
Shelf Life
Shelf life depends on various factors including storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized forms have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is essential for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing.
The tag type is determined during production. If you require a specific tag, please inform us; we will prioritize its development.
Synonyms
SET1; CAGL0L12980g; Histone-lysine N-methyltransferase; H3 lysine-4 specific; EC 2.1.1.354; COMPASS component SET1; SET domain-containing protein 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
Candida glabrata (strain ATCC 2001 / CBS 138 / JCM 3761 / NBRC 0622 / NRRL Y-65) (Yeast) (Torulopsis glabrata)
Target Names
SET1
Uniprot No.

Target Background

Function
Catalytic component of the COMPASS (Set1C) complex. Specifically mono-, di-, and trimethylates histone H3, forming H3K4me1/2/3, which plays a critical role in telomere length maintenance and transcription elongation regulation.
Database Links
Protein Families
Class V-like SAM-binding methyltransferase superfamily
Subcellular Location
Nucleus. Chromosome.

Q&A

What is the function of SET1 in Candida glabrata?

SET1 in C. glabrata functions as the primary histone methyltransferase responsible for mono-, di-, and trimethylation of histone H3 at lysine 4 (H3K4). As evidenced by deletion studies, Set1 is the sole H3K4 methyltransferase in C. glabrata under log-phase growth conditions, since deletion of SET1 abolishes all forms of H3K4 methylation . The protein contains a highly conserved SET domain that is critical for its catalytic activity, with the H1048K mutation rendering the enzyme catalytically inactive . SET1 plays a particularly important role in regulating azole-induced gene expression, especially genes involved in ergosterol biosynthesis, contributing to intrinsic drug resistance .

How does C. glabrata SET1 differ from orthologous proteins in other fungal species?

While SET1 maintains its fundamental role as an H3K4 methyltransferase across fungal species, significant functional differences exist:

SpeciesEffect of SET1 deletion on azole susceptibilityPrimary mechanismEffect on virulence
C. glabrataIncreased susceptibilityReduced ERG gene expressionPotentially reduced (inferred)
S. cerevisiaeIncreased susceptibilityDecreased efflux pump expressionNot applicable
C. albicansNo significant changeNot applicableReduced

In C. glabrata, Set1 governs azole resistance by epigenetically regulating ERG gene expression without affecting drug efflux systems . In contrast, SET1 deletion in S. cerevisiae decreases resistance by reducing expression and function of the efflux pump Pdr5 . Intriguingly, loss of SET1 in C. albicans alters virulence but does not affect azole sensitivity, suggesting divergent evolutionary paths in epigenetic regulation . These differences may relate to sterol uptake capabilities, as C. glabrata and S. cerevisiae can uptake sterols under various conditions, while C. albicans cannot .

What methodological approaches are used to generate and validate SET1 deletion mutants?

Creating reliable SET1 deletion mutants requires a systematic approach:

  • Gene deletion strategy: Using homologous recombination with a selectable marker (typically URA3 in auxotrophic strains like C. glabrata 2001HTU) . PCR-amplified cassettes containing the marker flanked by homologous sequences upstream and downstream of SET1 are transformed into cells.

  • Complementation verification: Cloning the full SET1 locus (including promoter, 5'UTR, coding sequence, and 3'UTR) into a plasmid like pGRB2.0 and transforming it back into the deletion strain confirms phenotype reversibility .

  • Site-directed mutagenesis: Creating catalytically inactive SET1 (H1048K mutation) through site-directed mutagenesis serves as a critical control, demonstrating that methyltransferase activity specifically mediates the observed phenotypes .

  • Validation techniques:

    • Western blotting with H3K4me1, H3K4me2, and H3K4me3 antibodies to confirm loss of methylation

    • Growth assays on azole-containing media to assess drug susceptibility

    • RNA-seq to evaluate transcriptional consequences

    • Chromatin immunoprecipitation to analyze histone modification patterns

How does one effectively measure azole susceptibility changes in SET1 mutants?

Measuring azole susceptibility requires multiple complementary approaches:

  • Spot dilution assays: Serial dilutions of yeast cultures (typically 10-fold) spotted on solid media containing increasing concentrations of azole drugs. This method effectively visualizes growth differences between wild-type and SET1 mutant strains across a concentration gradient .

  • Broth microdilution: Determining minimum inhibitory concentration (MIC) values in liquid culture according to Clinical and Laboratory Standards Institute (CLSI) guidelines. This quantitative approach provides standardized susceptibility measurements.

  • Growth curve analysis: Monitoring growth kinetics in liquid media containing sub-MIC concentrations of azoles using microplate readers to detect subtle differences in growth rates and lag phases.

  • Time-kill assays: Assessing cell viability at different timepoints after azole exposure to determine whether SET1 deletion affects fungicidal versus fungistatic drug responses.

  • Checkerboard assays: Testing combinations of azoles with other antifungals to evaluate whether SET1 deletion alters drug interaction profiles (synergy, antagonism, or indifference) .

How does Set1-mediated H3K4 methylation specifically regulate ERG gene expression during azole stress?

The regulation of ERG genes by Set1 involves a sophisticated epigenetic mechanism:

  • Basal versus induced regulation: SET1 deletion in C. glabrata does not significantly alter ERG gene expression under untreated conditions but prevents their proper induction upon azole treatment. This contrasts with S. cerevisiae, where SET1 affects basal ERG gene expression .

  • Chromatin dynamics: Chromatin immunoprecipitation analysis reveals that H3K4 trimethylation is present on actively transcribed ERG gene chromatin and increases upon azole induction. This increase depends on Set1's catalytic activity, as demonstrated by the H1048K mutant .

  • Transcriptional coverage: Set1 regulates all 12 genes involved in the late ergosterol biosynthesis pathway, including the critical azole target ERG11 and the downstream gene ERG3 . This coordinated regulation suggests that Set1 functions within a broader regulatory network controlling ergosterol homeostasis.

  • Mechanistic specificity: Unlike in S. cerevisiae, C. glabrata SET1 deletion does not alter expression of drug efflux pumps (CDR1) or their regulator (PDR1), indicating a specific effect on ergosterol biosynthesis rather than general stress response pathways .

What are the experimental challenges in studying histone modifications in clinical isolates of C. glabrata?

Investigating histone modifications in clinical isolates presents several methodological challenges:

  • Genetic heterogeneity: Clinical isolates often display significant genetic diversity, complicating the interpretation of epigenetic data. Researchers must account for strain-specific variations that might influence histone modification patterns independently of drug resistance mechanisms.

  • Technical standardization: ChIP protocols optimized for laboratory strains may require significant adaptation for clinical isolates due to differences in cell wall composition, chromatin accessibility, and antibody specificity.

  • Comparative controls: Establishing appropriate control strains for clinical isolates is challenging. Ideally, researchers should obtain matched susceptible/resistant isolates from the same patient before and after treatment failure.

  • Integration with other data types: Correlating histone modification patterns with transcriptomic, genomic, and phenotypic data requires sophisticated multi-omics approaches and statistical methods to identify causal relationships.

  • Sample limitations: Clinical samples often provide limited material, making it difficult to perform comprehensive ChIP-seq analyses that require substantial input material. Adapting low-input ChIP protocols becomes essential .

How do the COMPASS complex components interact with Set1 to influence drug resistance?

The COMPASS complex components play distinct roles in mediating Set1 function and drug resistance:

ComponentEffect on H3K4 methylation in C. glabrataImpact on azole susceptibilityComparison to S. cerevisiae
Set1Required for all H3K4me1/2/3Major increase when deletedSimilar effect
Swd1Required for all H3K4me1/2/3Similar to Set1 deletionSimilar effect
Bre2Required for all H3K4me1/2/3Similar to Set1 deletionSimilar effect
Spp1Complete loss of H3K4me3, reduced H3K4me1/2Intermediate phenotypeDifferent: only H3K4me3 affected in S. cerevisiae

What is the relationship between SET1-dependent regulation and other known antifungal resistance mechanisms?

SET1-dependent epigenetic regulation interacts with established resistance mechanisms in complex ways:

  • Ergosterol pathway mutations: While SET1 regulates ERG gene expression, it operates independently from mutations in ERG genes that commonly cause resistance. SET1 deletion would likely still increase azole susceptibility in strains with ERG gene mutations by preventing compensatory upregulation of other pathway components.

  • Efflux pump overexpression: In C. glabrata, SET1 deletion does not affect expression or function of the major efflux pump CDR1 or its regulator PDR1, suggesting parallel rather than sequential pathways . This contrasts with S. cerevisiae, where SET1 affects efflux pump expression .

  • Sterol uptake mechanisms: SET1 may interact with sterol uptake pathways, as slight increases in AUS1 transcript levels (encoding a sterol transporter) are observed in SET1-deleted C. glabrata under untreated conditions . This suggests potential compensatory mechanisms when ergosterol synthesis is compromised.

  • Clinical relevance: Clinical isolates lacking SET1 show hypersusceptibility to azoles attributed to reduced ERG11 expression rather than defects in drug efflux, confirming the importance of this mechanism in clinical settings .

What are the optimal approaches to studying Set1 catalytic activity in vitro?

Establishing robust in vitro assays for Set1 methyltransferase activity requires careful consideration:

  • Protein expression systems: Recombinant expression of C. glabrata Set1 presents challenges due to its large size and multiple domains. Recommended approaches include:

    • Baculovirus expression systems for full-length protein

    • Bacterial expression of the isolated SET domain for basic catalytic studies

    • Co-expression with other COMPASS components for complex reconstitution

  • Substrate preparation: Using recombinant histone H3, nucleosomes, or synthetic H3 tail peptides as substrates. Nucleosomes typically provide more physiologically relevant results than histone peptides.

  • Activity assays: Several complementary methods can measure methyltransferase activity:

    • Radiometric assays using tritiated S-adenosyl methionine (SAM)

    • Antibody-based detection of methylated products via Western blotting

    • Mass spectrometry to precisely quantify mono-, di-, and trimethylation states

    • Fluorescence-based assays measuring SAM conversion to SAH

  • Inhibitor screening: In vitro assays can evaluate potential Set1 inhibitors by:

    • Determining IC50 values for candidate compounds

    • Assessing selectivity against human SET-domain proteins

    • Characterizing inhibition mechanisms (competitive vs. non-competitive)

How should researchers design experiments to distinguish direct from indirect Set1 effects?

Differentiating direct from indirect effects of Set1 requires sophisticated experimental designs:

  • Temporal analysis: Time-course experiments examining the order of events following azole exposure can help establish causality. If H3K4 methylation changes precede transcriptional changes, direct regulation is more likely.

  • ChIP-seq and CUT&RUN approaches: These techniques can map Set1 occupancy and H3K4 methylation patterns genome-wide, identifying direct targets where both Set1 binding and increased methylation occur upon azole treatment .

  • Targeted mutations: Mutating predicted Set1 binding sites in ERG gene promoters would prevent direct regulation while leaving indirect pathways intact.

  • Rapid depletion systems: Using auxin-inducible degron (AID) tags to rapidly deplete Set1 can help distinguish immediate (likely direct) from delayed (likely indirect) effects on gene expression.

  • Transcription factor interactions: Examining whether Set1-dependent H3K4 methylation affects the recruitment or activity of known ERG gene transcription factors like Upc2 can clarify the regulatory mechanism .

What methodological approaches best characterize the dynamics of H3K4 methylation during azole stress?

Capturing the dynamic nature of H3K4 methylation during azole stress requires specialized approaches:

  • Time-resolved ChIP-seq: Performing ChIP for H3K4me1/2/3 at multiple timepoints after azole exposure (e.g., 15 min, 30 min, 1 hour, 2 hours, 4 hours) to track methylation changes preceding and during ERG gene induction .

  • Sequential ChIP (Re-ChIP): This technique can determine whether different methylation states (mono-, di-, tri-) occur sequentially or independently on the same histone molecules during the stress response.

  • Single-cell epigenomics: Techniques like single-cell CUT&Tag can reveal cell-to-cell variation in methylation patterns, identifying potential resistant subpopulations with distinct epigenetic profiles.

  • Live-cell imaging: Though technically challenging, systems using fluorescently tagged histone readers specific for different methylation states could visualize methylation dynamics in real-time during azole treatment.

  • Integration with transcriptional data: Correlating H3K4 methylation changes with nascent transcription (e.g., using NET-seq or TT-seq) rather than steady-state mRNA levels provides better temporal resolution of the regulatory relationship .

How might Set1 inhibition be developed as an adjunctive antifungal strategy?

Developing Set1 inhibitors as adjunctive therapies presents both opportunities and challenges:

  • Target validation: The increased azole susceptibility in SET1 deletion mutants provides strong validation for Set1 as a potential drug target. Particularly promising is evidence that clinical isolates lacking SET1 also show hypersusceptibility to azoles .

  • Inhibitor design approaches:

    • Structure-based design targeting the SET domain catalytic site

    • Peptidomimetics disrupting COMPASS complex assembly

    • Allosteric inhibitors affecting conformational changes required for activity

    • Targeted protein degradation approaches (PROTACs) specific for fungal Set1

  • Combination therapy potential: Set1 inhibitors could synergize with azoles by preventing adaptive ERG gene upregulation, potentially overcoming common resistance mechanisms. This might allow for reduced azole dosing, minimizing toxicity issues.

  • Selective toxicity considerations: The challenge lies in developing compounds that selectively inhibit fungal Set1 without affecting human SET-domain proteins (like SET1A/B, MLL1-4). Exploiting structural differences in the SET domain or in complex formation may provide selectivity.

  • Delivery systems: Developing appropriate formulations for antifungal combination therapy, potentially including nanoparticle-based delivery systems that could co-deliver azoles and Set1 inhibitors.

What are the broader implications of epigenetic regulation for fungal adaptation to host environments?

Epigenetic regulation likely plays fundamental roles in fungal adaptation beyond drug resistance:

  • Niche adaptation: Set1-mediated H3K4 methylation may regulate genes involved in adapting to specific host microenvironments, potentially explaining why SET1 deletion reduces virulence in C. albicans .

  • Immune evasion: Epigenetic mechanisms could regulate the expression of cell wall components and secreted factors that interact with host immune cells, potentially enabling rapid adaptation to immune surveillance.

  • Biofilm formation: Epigenetic regulation might contribute to the phenotypic heterogeneity observed in biofilms, with potential implications for treatment of biofilm-associated infections.

  • Host-pathogen interactions: The potential crosstalk between host and pathogen epigenetic machinery represents an unexplored frontier, as host cells also use histone modifications to regulate immune responses.

  • Stress memory: Epigenetic modifications could provide a form of cellular memory allowing more rapid adaptation to previously encountered stresses, including antifungal drugs or host defense mechanisms .

What novel experimental systems could advance our understanding of Set1 function in pathogenic contexts?

Innovative experimental approaches could significantly advance the field:

  • Ex vivo infection models: Developing complex tissue culture systems that better mimic host environments while allowing molecular analysis of Set1 function during infection processes.

  • CRISPR-based epigenetic modulators: Using catalytically inactive Cas9 fused to histone demethylases could provide targeted reversal of H3K4 methylation at specific genes, allowing precise dissection of Set1's gene-specific functions.

  • In vivo real-time monitoring: Developing reporter systems that reflect Set1 activity or H3K4 methylation status in live cells during infection, possibly using methylation-specific reader domains fused to fluorescent proteins.

  • Organoid infection models: Utilizing human organoids to study C. glabrata infections in more physiologically relevant contexts, combining this with single-cell transcriptomics and epigenomics.

  • Patient-derived xenograft models: Studying clinical isolates in immunocompromised mouse models to correlate Set1 function with in vivo drug responses and tissue tropism.

  • Multi-species infection models: Examining how Set1-mediated regulation functions during polymicrobial infections, which better represent many clinical scenarios .

What statistical approaches are most appropriate for analyzing SET1-dependent chromatin modifications?

Analyzing SET1-dependent chromatin modifications requires sophisticated statistical methods:

  • Differential binding analysis: Tools like DiffBind or DESeq2 can identify genomic regions with statistically significant changes in H3K4 methylation between conditions (untreated vs. azole-treated) or genotypes (WT vs. set1Δ).

  • Integration of multiple histone marks: Multivariate approaches can analyze the interdependence of different histone modifications (H3K4me1/2/3, H3K27ac, etc.) in response to SET1 deletion or azole treatment.

  • Peak calling optimization: Standard ChIP-seq peak callers may require parameter optimization for the broad H3K4me3 peaks characteristic of actively transcribed genes in yeasts.

  • Batch effect correction: Methods like ComBat or RUVseq are essential when comparing ChIP-seq data across multiple experiments, particularly for clinical isolates.

  • Time-series analysis: For time-course experiments, tools designed for temporal data (e.g., ImpulseDE2) can identify significant changes in methylation dynamics rather than static differences at individual timepoints.

  • Correlation with expression data: Beyond simple correlation analysis, more sophisticated approaches like BETA or EPIC can integrate ChIP-seq with RNA-seq to identify functional relationships between methylation changes and transcriptional outcomes .

How should researchers interpret seemingly contradictory data across different Candida species?

Resolving apparent contradictions in SET1 function across Candida species requires careful consideration:

  • Evolutionary context: Phylogenetic analysis of SET1 and related genes across species can provide context for functional divergence. C. glabrata is more closely related to S. cerevisiae than to C. albicans, potentially explaining some functional similarities .

  • Methodological standardization: Ensuring comparable experimental conditions, genetic backgrounds, and analytical methods when comparing across species. Different azole concentrations or exposure times might explain some disparities.

  • Compensatory mechanisms: Species-specific compensatory pathways might mask Set1 functions in some contexts. For example, C. albicans' limited sterol uptake capacity compared to C. glabrata might explain different dependencies on SET1 for azole resistance .

  • Functional redundancy: Examining potential redundancy with other histone methyltransferases or chromatin modifiers that might be species-specific.

  • Contextual specificity: Carefully defining the precise phenotypes being compared, as SET1 deletion might affect different aspects of azole response (e.g., immediate survival vs. adaptation) across species.

  • Validation approaches: Using complementation experiments where SET1 from one species is expressed in the deletion mutant of another species to directly test functional conservation .

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.