Recombinant Candida glabrata Helicase SWR1 (SWR1), partial

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder

Note: We will prioritize shipping the format currently in stock. If you have specific format requirements, please indicate them during order placement for custom preparation.

Lead Time
Delivery times vary depending on the purchase method and location. Please consult your local distributor for precise delivery estimates.

Note: All proteins are shipped with standard blue ice packs unless otherwise specified. 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. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our standard glycerol concentration is 50%, which can serve as a guideline.
Shelf Life
Shelf life depends on various factors including storage conditions, buffer composition, temperature, and the protein's inherent 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
The tag type will be determined during the manufacturing process.

Tag type is determined during production. If you require a specific tag, please inform us; we will prioritize its development.

Synonyms
SWR1; CAGL0M01188g; Helicase SWR1; EC 3.6.4.12
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
SWR1
Uniprot No.

Target Background

Function

Catalytic component of the SWR1 complex. This complex mediates the ATP-dependent exchange of histone H2A for the H2A variant HZT1, thereby regulating the transcription of specific genes through chromatin remodeling.

Database Links
Protein Families
SNF2/RAD54 helicase family, SWR1 subfamily
Subcellular Location
Nucleus.

Q&A

What is the primary function of Candida glabrata SWR1 helicase in chromatin remodeling?

SWR1 helicase in C. glabrata functions as a critical component of the SWR1 chromatin remodeling complex, which catalyzes the ATP-dependent exchange of canonical histone H2A with the histone variant H2A.Z at specific chromosomal locations. This process is essential for regulating transcription, DNA repair, and maintaining genome integrity. Within the fungal pathogen C. glabrata, SWR1 modulates chromatin structure in response to environmental changes and stressors, playing a pivotal role in gene expression regulation that may impact pathogenicity and drug resistance mechanisms .

What are the optimal storage and handling conditions for maintaining recombinant C. glabrata SWR1 stability?

For optimal stability of recombinant C. glabrata Helicase SWR1, storage conditions should be carefully controlled. The lyophilized form maintains stability for up to 12 months at -20°C to -80°C, while the liquid form remains stable for approximately 6 months under the same temperature conditions . For working protocols, it's advisable to reconstitute the protein in deionized sterile water to a concentration of 0.1-1.0 mg/mL with the addition of 5-50% glycerol (final concentration). The default recommendation is 50% glycerol for long-term storage . Notably, repeated freeze-thaw cycles significantly compromise protein integrity and function, so working aliquots should be stored at 4°C for no longer than one week . For experimental use, centrifuging the vial briefly before opening is recommended to ensure all contents settle at the bottom.

How can researchers effectively validate the functional activity of recombinant SWR1 helicase in vitro?

Validating the functional activity of recombinant C. glabrata SWR1 helicase requires a multi-faceted approach. Researchers should:

  • Assess ATP hydrolysis activity: Measure ATP hydrolysis rates using colorimetric phosphate detection assays. Active SWR1 should demonstrate DNA-stimulated ATPase activity.

  • Verify DNA binding capacity: Perform electrophoretic mobility shift assays (EMSA) to confirm the protein's ability to bind DNA substrates.

  • Examine histone exchange activity: Use reconstituted nucleosome substrates and fluorescently labeled H2A.Z to monitor the exchange of H2A for H2A.Z, which can be analyzed via gel-based assays or fluorescence techniques.

  • Confirm complex formation: If studying in the context of the full SWR1 complex, validate interactions with other complex components through co-immunoprecipitation or size-exclusion chromatography.

The protein's functional integrity can be compromised by improper handling, so careful attention to temperature, buffer composition, and the presence of stabilizing factors like glycerol is essential for maintaining enzymatic activity . Researchers should establish baseline activity parameters for each new protein preparation to ensure consistency across experiments.

How does SWR1 contribute to azole drug resistance mechanisms in C. glabrata?

SWR1 contributes to azole drug resistance in C. glabrata through its role in chromatin remodeling and subsequent epigenetic regulation of genes involved in ergosterol biosynthesis and drug efflux. Research indicates that SWR1-mediated incorporation of histone variant H2A.Z at specific genomic loci may facilitate the transcriptional regulation of azole resistance genes . This process appears to work in coordination with other epigenetic modulators, particularly the histone methyltransferase Set1 .

Set1-mediated histone H3K4 methylation has been demonstrated to govern intrinsic drug resistance in C. glabrata through epigenetic control of azole-induced ERG gene expression . When SWR1 complex subunits (such as Swd1) are deleted along with SET1, there is a notable loss of histone H3K4 methylation, which correlates with increased susceptibility to azole drugs . This suggests a cooperative relationship between the SWR1 complex and Set1-dependent histone modifications in maintaining the expression of genes critical for azole resistance.

Recent research has also revealed that zinc cluster transcription factors like Hap1A (Zcf4) and Hap1B (Zcf27) directly regulate ERG genes crucial for azole resistance, with deletion of HAP1B/ZCF27 resulting in increased azole susceptibility . These transcription factors likely operate within the chromatin landscape established by SWR1 and other chromatin remodelers.

What is known about the interaction between SWR1 and the NuA4 histone acetyltransferase complex in Candida species?

In Candida species, the interaction between SWR1 and NuA4 histone acetyltransferase complexes represents a sophisticated mechanism of chromatin regulation that impacts cell fate determination. Unlike in Saccharomyces cerevisiae where these complexes remain relatively distinct, C. albicans (and likely C. glabrata) exhibits a dynamic merge and separation of these complexes depending on cellular state .

Key aspects of this interaction include:

  • State-dependent association: NuA4 and SWR1 merge together in the yeast state but separate into two distinct complexes in the hyphal state of C. albicans . This dynamic association may be conserved in polymorphic fungi including C. glabrata.

  • Acetylation-dependent regulation: The interaction is controlled by acetylation of Eaf1 K173, specifically recognized by the YEATS domain of Yaf9 . This acetylation serves as a molecular switch for complex association.

  • Enzymatic control: The reversible acetylation and deacetylation of Eaf1 by the acetyltransferase Esa1 and the deacetylase Hda1 regulate the merge and separation of these complexes .

  • Nutritional signaling: The regulatory mechanism is triggered by Brg1 recruitment of Hda1 to chromatin in response to nutritional signals .

  • Coordinated chromatin association: Orchestrated promoter association of Esa1, Hda1, Swr1, and H2A.Z occurs during yeast-hyphae transitions .

This dynamic relationship between SWR1 and NuA4 complexes represents an evolved mechanism that likely allows Candida species to rapidly respond to environmental changes, including host environments and antifungal stressors. The interaction may be particularly important for regulating genes involved in morphogenesis, metabolism, and drug resistance .

How can CRISPR-Cas9 genome editing be optimized for studying SWR1 function in C. glabrata?

Optimizing CRISPR-Cas9 genome editing for studying SWR1 function in C. glabrata requires addressing several unique challenges related to this pathogen's biology. A comprehensive approach should include:

  • Efficient delivery system: C. glabrata has a rigid cell wall that complicates transformation. Using electroporation with optimized parameters (1.5 kV, 200 Ω, 25 μF) can improve transformation efficiency for CRISPR components. Lithium acetate transformation protocols specifically adapted for C. glabrata work well when coupled with cell wall weakening agents like β-mercaptoethanol.

  • Codon-optimized Cas9: Using a Candida-optimized Cas9 expression construct driven by a strong, constitutive promoter like PGK1 enhances editing efficiency. For SWR1 studies, the expression level of Cas9 should be carefully controlled to avoid growth defects that might confound SWR1 phenotypes.

  • Guide RNA design: For targeting SWR1 or its interacting partners, design sgRNAs with high on-target and low off-target scores. Focus on:

    • Targeting conserved helicase domains for null mutations

    • Creating precise mutations in ATP-binding sites to study catalytic requirements

    • Introducing epitope tags for protein interaction studies

    • The RNA polymerase III promoter (SNR52) has proven effective for sgRNA expression in C. glabrata

  • Homology-directed repair templates: Design with longer homology arms (>500 bp) than typically used in S. cerevisiae. For precise SWR1 domain analysis, incorporate silent mutations in the PAM site or sgRNA target sequence to prevent re-cutting after successful editing.

  • Selection strategies: For SWR1 studies, direct selection for edited cells may be challenging as SWR1 mutations can affect chromatin structure and potentially cell viability. Using marker-free editing with transient selection can help avoid potential artifacts from selection markers in chromatin studies.

  • Validation approaches: Confirm edits using a multi-modal approach:

    • PCR and sequencing for genetic confirmation

    • Western blotting to verify protein expression changes

    • ChIP-seq to assess altered H2A.Z deposition patterns in SWR1 mutants

    • RNA-seq to determine transcriptional consequences

    • Drug susceptibility testing to evaluate azole resistance phenotypes

This optimized CRISPR-Cas9 approach enables precise genetic manipulation of SWR1 and its interacting partners, facilitating detailed structure-function analyses and uncovering the role of specific SWR1 domains in chromatin remodeling and drug resistance mechanisms .

What approaches can be used to investigate the interplay between SWR1-mediated chromatin remodeling and transcription factor activity in azole resistance?

Investigating the interplay between SWR1-mediated chromatin remodeling and transcription factor activity in azole resistance requires an integrated approach combining genetic, genomic, and biochemical techniques:

  • Sequential ChIP (ChIP-reChIP) analysis: This technique can detect co-occupancy of SWR1 components and transcription factors like Pdr1, Hap1A/B (Zcf4/Zcf27), and Upc2 at promoters of azole resistance genes. This approach has successfully demonstrated direct regulation of ERG genes by zinc cluster transcription factors in C. glabrata .

  • CRISPR interference (CRISPRi)-based genetic dissection: Deploy a nuclease-dead Cas9 (dCas9) system to reversibly repress either SWR1 components or specific transcription factors, allowing time-resolved analysis of their interdependence. This approach can determine whether SWR1 activity is prerequisite for transcription factor binding or vice versa.

  • Inducible degron systems: Apply auxin-inducible or temperature-sensitive degron tags to SWR1 complex components to achieve rapid protein depletion, enabling analysis of immediate transcription factor binding changes before secondary effects emerge.

  • Combinatorial genetic analysis: Generate strains with various combinations of mutations in SWR1 complex components and transcription factors (e.g., swr1Δ/hap1bΔ double mutants) to quantify epistatic relationships. Research has shown that deletion of HAP1B/ZCF27 results in azole hypersusceptibility through decreased expression of ERG genes .

  • Nucleosome positioning analysis: Compare MNase-seq profiles between wild-type and SWR1-deficient strains to map changes in chromatin accessibility at transcription factor binding sites. This can reveal whether SWR1-dependent H2A.Z deposition is required for transcription factor access to resistance gene promoters.

  • Histone modification mapping: Employ ChIP-seq for histone marks (particularly H3K4 methylation) in conjunction with transcription factor binding and gene expression analysis. Studies have shown that Set1-mediated H3K4 methylation, which may cooperate with SWR1 activity, is required for proper azole-induced ERG gene expression .

  • Proteomic approaches: Use proximity-labeling techniques (BioID or APEX) with either SWR1 components or transcription factors as baits to identify protein interaction networks in drug-naive versus azole-exposed conditions.

  • In vitro nucleosome remodeling assays: Reconstitute core components of the SWR1 complex with recombinant proteins and assess how transcription factors influence H2A.Z deposition activity on nucleosome templates containing relevant promoter sequences.

This multi-faceted approach has revealed that transcription factors like Hap1B directly regulate ERG genes involved in ergosterol biosynthesis, while SWR1-mediated chromatin remodeling appears to establish the appropriate chromatin landscape for this regulation . The data suggests a model where SWR1 complex activity and histone modifications work cooperatively with specific transcription factors to enable rapid and sustained transcriptional responses to azole exposure.

How have SWR1 complex components evolved between non-pathogenic yeasts and pathogenic Candida species?

The evolution of SWR1 complex components between non-pathogenic yeasts like Saccharomyces cerevisiae and pathogenic Candida species reveals significant adaptations that may contribute to virulence and stress response capabilities. Comparative analysis shows:

  • Functional specialization: While the core enzymatic function of SWR1 in histone variant exchange is conserved, pathogenic Candida species exhibit specialized regulatory mechanisms. Most notably, C. glabrata and C. albicans demonstrate dynamic association and dissociation between the SWR1 and NuA4 complexes in response to environmental conditions, a feature not observed in S. cerevisiae . This adaptability likely represents an evolutionary advantage for survival in diverse host environments.

  • Subunit composition differences: Though many SWR1 complex subunits are conserved across fungal species, pathogenic Candida demonstrate subtle but important differences. For instance, the functional outcomes of deleting supposedly homologous subunits can differ between species. Deletion of SPP1 in C. glabrata abolishes H3K4 trimethylation and significantly reduces mono- and dimethylation, whereas in S. cerevisiae, spp1Δ only disrupts trimethylation while maintaining normal levels of mono- and dimethylation . These differences suggest altered complex assembly or subunit function.

  • Regulatory network integration: In C. glabrata, SWR1 complex activity appears more tightly integrated with stress response pathways, particularly those involved in azole resistance. The complex works in concert with transcription factors like Hap1A/B (Zcf4/Zcf27) that have undergone duplication and functional divergence following whole genome duplication events . This integration reflects evolutionary adaptation to antifungal pressure.

  • Post-translational regulation: The mechanisms controlling SWR1 complex activity have diverged, with C. albicans showing acetylation-dependent regulation of complex assembly mediated by the YEATS domain of Yaf9 recognizing acetylated Eaf1 . This represents a specialized control mechanism potentially allowing more rapid responses to environmental changes.

  • Gene duplication and neo-functionalization: Following whole genome duplication, C. glabrata has retained and repurposed duplicated genes that interact with chromatin remodeling pathways. For example, the HAP1 gene underwent duplication resulting in HAP1A and HAP1B (ZCF4 and ZCF27), which now serve distinct functions in regulating ERG genes under different conditions, potentially in coordination with chromatin remodelers like SWR1 .

These evolutionary adaptations in SWR1 complex composition and regulation likely contribute to the enhanced stress tolerance and host adaptation capabilities of pathogenic Candida species, representing important targets for understanding pathogenicity mechanisms and developing novel antifungal strategies.

What can phylogenetic analysis of SWR1 helicase domains tell us about functional specialization in fungal pathogens?

This evolutionary perspective on SWR1 helicase domains provides a framework for understanding how chromatin remodeling machinery has been adapted in fungal pathogens to enhance survival in host environments and resist antifungal therapies. It also highlights potential functional regions that could be targeted for the development of novel antifungal approaches that specifically disrupt pathogen-adapted functions.

How might knowledge of SWR1 function be leveraged for developing novel diagnostic tools for Candida glabrata infections?

Knowledge of SWR1 function could be strategically leveraged to develop innovative diagnostic tools for Candida glabrata infections through several approaches:

  • SWR1-dependent chromatin signatures as biomarkers: The unique chromatin remodeling patterns mediated by SWR1 in C. glabrata create specific epigenetic signatures that could serve as diagnostic biomarkers. By developing antibodies or aptamers that recognize C. glabrata-specific histone modifications or H2A.Z deposition patterns, researchers could create highly specific immunodiagnostic assays .

  • Nucleic acid amplification targeting SWR1-regulated genes: Recent advances in rapid molecular diagnostics for C. glabrata have utilized recombinase-aided PCR methods targeting species-specific genes . Expanding these approaches to target unique SWR1-regulated genes could improve both sensitivity and specificity. For instance, the recently developed M1-mRAP assay, which combines recombinant human mannan-binding lectin beads with multiplex recombinase-aided PCR, could be adapted to target SWR1-regulated loci that are uniquely expressed during infection .

  • Protein-based detection systems: Antibodies raised against unique epitopes in C. glabrata SWR1 or its complex components could form the basis for lateral flow assays or ELISA-based diagnostics with improved species specificity. The high conservation of SWR1 within C. glabrata strains makes it an attractive target for species-specific detection .

  • SWR1-dependent drug resistance profiling: Given SWR1's role in azole resistance mechanisms, diagnostic tools that simultaneously detect C. glabrata and predict its likely drug resistance profile based on SWR1-associated biomarkers could guide early therapeutic decisions. Research has established connections between specific chromatin remodeling states and drug resistance phenotypes that could be exploited for this purpose .

  • Novel molecular beacon designs: Developing molecular beacons that specifically target unique sequences in the SWR1 gene or its regulated genes could enable rapid fluorescence-based detection systems. The internal transcribed spacer protein-2 (ITS2) gene has been successfully used in RPA-LFS (recombinase polymerase amplification-lateral flow strip) systems, and similar approaches could be adapted for SWR1-regulated genes .

  • Integrated diagnostic algorithms: Combining detection of SWR1-related biomarkers with other Candida-specific targets could create diagnostic algorithms with enhanced sensitivity and specificity. Recent research describing the novel C. glabrata protein Yhi1, which induces hyphal growth in C. albicans during mixed-species infections, suggests that multiple species-specific markers could be integrated into comprehensive diagnostic panels .

Implementation of these approaches could address the current limitations in diagnosing C. glabrata infections, such as the long turn-around time and low detection rate of traditional blood cultures. Molecular methods targeting SWR1-related biomarkers could potentially reduce detection time from days to hours, critical for timely intervention in these often drug-resistant infections .

What are the implications of SWR1-mediated chromatin remodeling for antifungal drug development and resistance management?

SWR1-mediated chromatin remodeling has significant implications for antifungal drug development and resistance management, offering both challenges and opportunities for next-generation therapeutic approaches:

  • Novel therapeutic targets in chromatin pathways: SWR1 and its associated chromatin remodeling machinery represent potential targets for novel antifungal drugs. Inhibiting SWR1 complex activity could sensitize C. glabrata to existing azole drugs by preventing the chromatin remodeling necessary for activating drug resistance genes. Research has shown that disruption of epigenetic mechanisms, such as Set1-mediated histone H3K4 methylation, increases azole susceptibility, suggesting similar vulnerabilities in SWR1-dependent pathways .

  • Combination therapy strategies: Targeting SWR1-dependent chromatin remodeling alongside traditional antifungal drugs could create synergistic effects. For example, compounds that disrupt the interaction between SWR1 and transcription factors like Hap1B/Zcf27, which directly regulate ergosterol biosynthesis genes, might prevent adaptive resistance mechanisms from activating . This approach could potentially restore susceptibility to azoles in resistant strains.

  • Pre-emptive resistance management: Understanding the SWR1-dependent chromatin changes that precede full resistance development could enable early intervention strategies. Research has shown that C. glabrata undergoes significant chromatin reorganization as part of its response to azole exposure, before developing stable genetic mutations in regulators like Pdr1 . Monitoring these early chromatin signatures could guide timely adjustment of treatment regimens.

  • Epigenetic biomarkers of resistance potential: The SWR1-mediated H2A.Z deposition patterns and associated histone modifications could serve as biomarkers for predicting a strain's propensity to develop resistance. This could inform personalized treatment approaches based on the epigenetic profile of the infecting strain, potentially identifying high-risk infections that warrant more aggressive initial therapy or closer monitoring.

  • Targeting cooperative transcription networks: SWR1 works in concert with specific transcription factors to regulate resistance genes. Research has revealed that zinc cluster transcription factors like Hap1A/B and Pdr1 depend on appropriate chromatin landscapes to function properly . Developing compounds that disrupt this cooperative relationship could provide selective pressure less likely to generate resistance through simple point mutations.

  • Exploiting species-specific adaptations: The evolutionary divergence in SWR1 complex regulation between pathogenic Candida and humans offers opportunities for selective targeting. The unique regulatory mechanisms governing the dynamic association between SWR1 and NuA4 complexes in Candida species represent a potentially fungal-specific vulnerability . Compounds that disrupt this dynamic, perhaps by targeting the YEATS domain of Yaf9 or the acetylation sites on Eaf1, could selectively impair fungal chromatin remodeling.

  • Addressing population heterogeneity: SWR1-mediated chromatin plasticity likely contributes to the rapid microevolution observed in clinical C. glabrata isolates . Therapeutic strategies that reduce this epigenetic plasticity might limit the ability of populations to develop heterogeneous resistance profiles, potentially slowing the emergence of resistant subpopulations during treatment.

These implications highlight the importance of considering chromatin-level regulation in both antifungal drug development and clinical management of resistant infections. By targeting the epigenetic machinery that enables adaptation to antifungal pressure, rather than just the end-point resistance mechanisms, novel therapeutic approaches could potentially overcome the persistent challenge of antifungal resistance in C. glabrata.

What are the major challenges in purifying functionally active recombinant SWR1 helicase, and how can they be addressed?

Purifying functionally active recombinant SWR1 helicase from C. glabrata presents several technical challenges that require strategic approaches to overcome:

  • Protein size and structural complexity

    • Challenge: Full-length SWR1 is a large protein with multiple domains that can complicate expression and folding.

    • Solution: Expression of partial constructs focusing on functional domains can improve yield and stability, as demonstrated by the commercial availability of partial SWR1 preparations . Systematic domain mapping using multiple truncation constructs can identify stable fragments that retain critical activities.

  • Expression system selection

    • Challenge: Different expression systems yield varying results for SWR1 quality and activity.

    • Solution: Both E. coli and Baculovirus expression systems have been successfully used , but with different outcomes. E. coli systems (product code CSB-EP739430CZI) provide higher yield but may lack post-translational modifications, while Baculovirus systems (product code CSB-BP739430CZI) better preserve native protein folding and modifications. For functional studies requiring ATP-dependent activities, the Baculovirus system often produces more consistently active preparations.

  • Protein solubility

    • Challenge: Helicases often have hydrophobic regions that promote aggregation during expression.

    • Solution: Incorporating solubility-enhancing tags (MBP, SUMO) can significantly improve solubility. Additionally, optimizing lysis and purification buffers with specific detergents (0.01-0.05% NP-40 or Triton X-100) and higher salt concentrations (300-500 mM NaCl) can maintain solubility without compromising activity.

  • Preserving ATP-dependent activity

    • Challenge: The ATPase activity critical for SWR1 helicase function is often lost during purification.

    • Solution: Including ATP analogs or ADP during purification can stabilize the nucleotide-binding domain. Maintaining a constant low concentration (0.2-0.5 mM) of ATP or non-hydrolyzable ATP analogs throughout purification has been shown to preserve helicase activity. Additionally, inclusion of 5-10% glycerol in all buffers helps maintain protein stability and enzymatic activity.

  • Co-factors and co-purification requirements

    • Challenge: SWR1 typically functions as part of a multi-protein complex, and isolation may disrupt necessary protein-protein interactions.

    • Solution: Co-expression with minimal functional partners or developing partial reconstitution strategies with separately purified subunits can restore activity. For specific studies, co-purification with critical interaction partners like Swc2 (which binds H2A.Z) can maintain biological relevance.

  • Post-purification stability

    • Challenge: Purified SWR1 helicase often demonstrates rapid activity loss during storage.

    • Solution: As indicated in the product datasheets, adding 5-50% glycerol (with 50% recommended as default) significantly improves stability . Additionally, flash-freezing in liquid nitrogen rather than slow freezing, and strict avoidance of freeze-thaw cycles by preparing single-use aliquots extends functional half-life. Storage at -80°C is preferred over -20°C for long-term activity preservation.

  • Functional validation methodology

    • Challenge: Assessing whether purified SWR1 retains biologically relevant activity.

    • Solution: Implementing a multi-tiered activity assessment approach:

      • Primary validation through ATP hydrolysis assays

      • Secondary validation via DNA/nucleosome binding assays

      • Tertiary validation through in vitro histone exchange assays with reconstituted nucleosomes

      • Quaternary validation via complementation of SWR1-deficient strains when possible

  • Batch-to-batch consistency

    • Challenge: Maintaining reproducible activity across different purification batches.

    • Solution: Developing standardized quality control metrics based on specific activity (activity units per mg protein) rather than just protein concentration or purity can ensure functional consistency. The >85% purity standard used for commercial preparations (as assessed by SDS-PAGE) provides a baseline, but functional assays are essential for confirming activity .

By systematically addressing these challenges, researchers can obtain functionally active SWR1 helicase preparations suitable for biochemical, structural, and functional studies, enabling deeper investigation of this protein's role in chromatin remodeling and antifungal resistance mechanisms.

What strategies can overcome the challenges in studying SWR1 complex dynamics in vivo within C. glabrata?

Studying SWR1 complex dynamics in vivo within C. glabrata presents unique challenges due to the organism's biology and technical limitations. The following strategies offer solutions to these challenges:

By integrating these approaches, researchers can overcome the technical challenges inherent to studying chromatin dynamics in C. glabrata, enabling deeper insights into how SWR1-mediated chromatin remodeling contributes to drug resistance and pathogenicity.

What are the most promising directions for studying the role of SWR1 in C. glabrata pathogenesis and host interaction?

The investigation of SWR1's role in C. glabrata pathogenesis and host interaction presents several promising research directions that could significantly advance our understanding of fungal virulence mechanisms:

  • SWR1-dependent regulation of host adaptation genes

    • Exploring how SWR1-mediated chromatin remodeling controls the expression of genes involved in host niche adaptation represents a frontier in understanding C. glabrata pathogenicity. Recent discoveries of C. glabrata-specific proteins like Yhi1, which facilitates interaction with C. albicans during mixed-species infections , suggest that SWR1 may regulate specialized virulence factors. Comprehensive ChIP-seq mapping of H2A.Z deposition across infection-relevant conditions could identify novel virulence gene clusters under SWR1 control.

  • SWR1 complex response to host immune signals

    • Investigating how host immune factors trigger chromatin remodeling through the SWR1 complex could reveal mechanisms of immune evasion. C. glabrata's remarkable ability to survive within phagocytes suggests sophisticated chromatin-level responses to oxidative stress that likely involve SWR1. Research has shown that C. glabrata has developed high resistance to reactive oxygen species, with only double mutants lacking both superoxide dismutase CgSod1 and the transcription factor CgYap1 being efficiently killed by macrophages . Exploring whether SWR1-dependent chromatin remodeling contributes to this stress resistance represents a promising research direction.

  • Dynamic regulation of SWR1-NuA4 association during infection

    • The dynamic assembly and disassembly of SWR1 and NuA4 complexes in response to environmental cues, as observed in C. albicans , may play crucial roles during host colonization and infection progression. Developing in vivo imaging techniques to track this dynamic in real-time during infection could provide unprecedented insights into how chromatin remodeling machinery responds to host environments.

  • SWR1-dependent epigenetic adaptation during chronic infection

    • Chronic C. glabrata infections often exhibit evolving drug resistance and virulence characteristics. Investigating whether SWR1-mediated chromatin remodeling contributes to this microevolution by creating epigenetic variants with enhanced survival capabilities could reveal new paradigms in pathogen adaptation. Recent studies of C. glabrata population genetics have revealed significant genetic diversity within the mitochondrial genome of clinical isolates , suggesting that nuclear-mitochondrial interactions, potentially involving chromatin remodeling, may impact virulence traits.

  • Interspecies chromatin dynamics in polymicrobial infections

    • C. glabrata often causes infections alongside other Candida species or bacteria. Exploring how SWR1-dependent chromatin states are altered during polymicrobial interactions could reveal mechanisms of synergistic pathogenicity. The recent discovery of the Yhi1 protein, which is secreted by C. glabrata to induce hyphal growth in C. albicans , demonstrates the importance of interspecies communication. Investigating whether SWR1 regulates genes involved in these interactions represents an exciting research frontier.

  • Tissue-specific chromatin adaptations

    • C. glabrata can colonize diverse host tissues with varying nutrient availability, oxygen levels, and immune surveillance. Characterizing how SWR1-dependent chromatin remodeling contributes to tissue-specific adaptations could identify targetable vulnerabilities. This direction could benefit from ex vivo infection models that more accurately recapitulate specific tissue microenvironments.

  • Integration of stress response signals at the chromatin level

    • Investigating how multiple stress signals (oxidative, osmotic, pH, nutrient limitation) are integrated through SWR1-dependent chromatin remodeling could reveal master regulatory mechanisms of pathogen adaptation. Research has demonstrated that C. glabrata employs zinc cluster transcription factors like Hap1A/B to regulate gene expression under different conditions , but how these transcription factors interact with the chromatin landscape established by SWR1 remains an open question.

  • Therapeutic targeting of SWR1-dependent chromatin states

    • Developing small molecule inhibitors that specifically disrupt SWR1 complex assembly or function in C. glabrata could provide novel antifungal approaches. Structure-guided design of compounds that interfere with ATP binding or protein-protein interactions within the complex could lead to pathogen-specific therapeutics that synergize with existing antifungals.

These research directions promise to uncover fundamental mechanisms by which chromatin remodeling contributes to fungal pathogenesis, potentially revealing novel therapeutic targets and diagnostic approaches for this increasingly important human pathogen.

How might emerging technologies like single-cell genomics and CRISPR screening advance our understanding of SWR1 function in chromatin remodeling?

Emerging technologies in single-cell genomics and CRISPR screening present transformative opportunities for understanding SWR1 function in chromatin remodeling, particularly within the context of fungal pathogens like C. glabrata. These cutting-edge approaches enable unprecedented resolution in studying chromatin dynamics and gene function:

  • Single-cell chromatin accessibility profiling

    • Technologies like scATAC-seq can reveal cell-to-cell heterogeneity in chromatin states within C. glabrata populations, potentially uncovering how SWR1-mediated H2A.Z deposition creates subpopulations with varying drug resistance or virulence properties. Recent studies have highlighted the importance of population heterogeneity in C. glabrata clinical isolates , but the epigenetic contribution to this heterogeneity remains largely unexplored. Applying single-cell chromatin profiling could reveal how SWR1 activity contributes to the emergence of resistant subpopulations during antifungal treatment.

  • Integrated single-cell multi-omics

    • Combined approaches that simultaneously profile chromatin accessibility, transcriptome, and proteome from the same cell can establish direct causal relationships between SWR1-dependent chromatin states and functional outcomes. These technologies could help resolve the temporal dynamics of how SWR1-mediated chromatin remodeling precedes and directs transcriptional responses to environmental changes, particularly during host-pathogen interactions.

  • High-resolution genome-wide CRISPR screening

    • Developing C. glabrata-optimized CRISPR interference (CRISPRi) or activation (CRISPRa) libraries targeting genes regulated by SWR1 could identify which target genes are most critical for specific phenotypes like drug resistance or virulence. This approach could help prioritize the extensive network of SWR1-regulated genes to identify the most promising therapeutic targets. Recent advances in CRISPR technology for fungal pathogens make this approach increasingly feasible.

  • Domain-focused CRISPR scanning

    • Using tiled CRISPR editing to introduce systematic mutations across the SWR1 gene could map functionally critical residues and domains with unprecedented resolution. This approach could identify subtle functional requirements beyond the known ATP-binding domains, potentially revealing novel regulatory interfaces specific to pathogenic fungi that could be exploited for drug development.

  • Temporal CRISPR perturbation

    • Inducible CRISPR systems that allow time-resolved disruption of SWR1 function could separate immediate from adaptive chromatin responses, revealing the primary targets and sequential remodeling events controlled by SWR1. This approach would be particularly valuable for understanding the rapid chromatin reorganization that occurs during antifungal drug exposure.

  • Single-molecule chromatin imaging

    • Emerging techniques like super-resolution microscopy coupled with live-cell DNA tracking can visualize individual SWR1 complexes as they engage with and remodel nucleosomes in real-time. This could reveal the kinetics and mechanistic details of how SWR1 selects target sites and executes H2A.Z deposition, particularly in response to specific environmental signals relevant to pathogenesis.

  • Spatial transcriptomics and chromatin mapping

    • Technologies that preserve spatial information while profiling gene expression and chromatin states could reveal how SWR1 activity varies across microenvironments during host colonization and biofilm formation. This approach could identify localized chromatin remodeling events that contribute to the structural and functional heterogeneity observed in C. glabrata biofilms, which are particularly resistant to antifungal treatments.

  • In situ chromatin conformation analysis

    • Techniques like Hi-C and its derivatives can map how SWR1-dependent changes in local chromatin structure influence higher-order genome organization and gene expression domains. This could reveal how chromatin remodeling coordinates the expression of functionally related genes, such as those involved in drug efflux or ergosterol biosynthesis, to mount coherent adaptive responses.

  • CRISPR-based synthetic genetic interaction mapping

    • Systematic genetic interaction screens using CRISPR technology could identify genes and pathways that buffer or enhance SWR1 function, revealing redundancy and compensation mechanisms that influence resistance phenotypes. This approach could identify combination therapeutic targets that create synthetic lethality when inhibited alongside SWR1-dependent pathways.

  • Long-read epigenetic sequencing

    • Technologies like nanopore sequencing that can simultaneously detect DNA sequence and modifications could provide integrated views of genetic variation and epigenetic states across SWR1 target loci. This approach could be particularly valuable for understanding how genetic variants in regulatory regions interact with SWR1-dependent chromatin remodeling to influence gene expression and phenotypic outcomes.

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.