Recombinant Saccharomyces cerevisiae COMPASS component SWD2 (SWD2)

Shipped with Ice Packs
In Stock

Description

Role in COMPASS and H3K4 Methylation

SWD2 is essential for maintaining COMPASS integrity and histone H3K4 methylation activity:

  • Set1 Stability: Depletion of SWD2 destabilizes the catalytic subunit Set1, reducing H3K4me2/me3 levels .

  • RNApII Recruitment: SWD2 cooperates with the N-terminal domain of Set1 (residues 1–200) to bind the phosphorylated C-terminal domain (CTD) of RNApII, facilitating COMPASS recruitment to transcription elongation complexes .

    • Deletion of Set1 residues 200–210 disrupts SWD2 binding and reduces H3K4 methylation .

    • A SWD2 F250A mutation in the WD40 domain impairs COMPASS assembly and H3K4me3 deposition .

Mechanistic Insights from Recombinant Studies

Key experiments using recombinant SWD2 and truncation mutants have elucidated its functional domains:

  • Yeast Two-Hybrid (Y2H) Assays: Demonstrated that SWD2 and Set1’s N-terminal domain form a composite interface for CTD binding. SWD2 deletion (swd2Δ) reduces CTD interaction by 70% .

  • In Vitro Reconstitution: Recombinant SWD2 F250A mutant incorporated into COMPASS shows reduced methyltransferase activity on nucleosomes compared to wild type .

  • Bypass Recruitment: Replacing Set1’s N-terminal domain with the Nrd1 CTD-interacting domain (CID) restores H3K4me3 independently of SWD2, confirming SWD2’s role in targeting .

Dual Role in Transcription Termination (APT Complex)

SWD2’s essentiality in S. cerevisiae stems from its dual functions:

  • APT Recruitment: SWD2 is required for APT-dependent termination of snoRNAs and mRNAs. Its WD40 domain interacts with Ref2, a termination factor .

  • Genetic Interactions: SWD2 depletion causes transcription readthrough defects, which are rescued by Ref2 overexpression or Sen1 fragments .

Evolutionary Conservation and Relevance

SWD2 homologs (e.g., Wdr82 in mammals) retain roles in H3K4 methylation:

  • Mammalian Systems: Wdr82 binds Setd1A and Ser5P-CTD, analogous to SWD2’s role in yeast COMPASS .

  • Fission Yeast: Schizosaccharomyces pombe has two SWD2 paralogs, each dedicated to COMPASS or APT, highlighting functional divergence .

Research Implications and Applications

Recombinant SWD2 studies have advanced understanding of:

  • Epigenetic Regulation: How COMPASS coordinates with transcription elongation to deposit H3K4me3.

  • Therapeutic Targets: WD40 domains in SWD2/Wdr82 are potential sites for modulating histone methylation in diseases like cancer .

Product Specs

Form
Lyophilized powder
Please note: We prioritize shipping the format currently in stock. However, if you have specific requirements for the format, please indicate them in your order notes. We will fulfill your request to the best of our ability.
Lead Time
Delivery time may vary depending on the purchasing method and location. For specific delivery information, please contact your local distributor.
Note: All proteins are shipped with standard blue ice packs by default. If you require dry ice shipping, please inform us in advance, as additional fees will apply.
Notes
Repeated freezing and thawing is not recommended. For optimal use, store working aliquots at 4°C for up to one week.
Reconstitution
We recommend briefly centrifuging the vial before opening to ensure the contents are settled at the bottom. Reconstitute the protein in deionized sterile water to a final concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% glycerol (final concentration) and aliquotting for long-term storage at -20°C/-80°C. Our standard final glycerol concentration is 50%, which can be used as a reference.
Shelf Life
Shelf life is influenced by various factors, including storage conditions, buffer composition, storage temperature, and the protein's inherent stability.
Generally, the shelf life of liquid form is 6 months at -20°C/-80°C. The shelf life of lyophilized form is 12 months at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt. Aliquoting is necessary for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
The tag type will be determined during the manufacturing process.
If you have a specific tag type in mind, please inform us, and we will prioritize the development of your specified tag.
Synonyms
SWD2; CPS35; SAF37; YKL018W; COMPASS component SWD2; Complex proteins associated with SET1 protein SWD2; Set1C component SWD2
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-329
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Saccharomyces cerevisiae (strain ATCC 204508 / S288c) (Baker's yeast)
Target Names
SWD2
Target Protein Sequence
MTTVSINKPN LLKFKHVKSF QPQEKDCGPV TSLNFDDNGQ FLLTSSSNDT MQLYSATNCK FLDTIASKKY GCHSAIFTHA QNECIYSSTM KNFDIKYLNL ETNQYLRYFS GHGALVNDLK MNPVNDTFLS SSYDESVRLW DLKISKPQVI IPSLVPNCIA YDPSGLVFAL GNPENFEIGL YNLKKIQEGP FLIIKINDAT FSQWNKLEFS NNGKYLLVGS SIGKHLIFDA FTGQQLFELI GTRAFPMREF LDSGSACFTP DGEFVLGTDY DGRIAIWNHS DSISNKVLRP QGFIPCVSHE TCPRSIAFNP KYSMFVTADE TVDFYVYDE
Uniprot No.

Target Background

Function
The COMPASS (Set1C) complex specifically mono-, di- and trimethylates histone H3 to form H3K4me1/2/3, which plays a role in telomere length maintenance and transcription elongation regulation. It is also involved in mediating RNA polymerase II termination. As a component of the cleavage and polyadenylation factor (CPF) complex, it plays a critical role in polyadenylation-dependent pre-mRNA 3'-end formation and cooperates with cleavage factors, including the CFIA complex and NAB4/CFIB. Additionally, it is a component of the APT complex, potentially involved in polyadenylation-independent transcript 3'-end formation.
Gene References Into Functions
  1. Swd2.2 counteracts condensin-mediated chromosome condensation by facilitating the function of the two CPF-associated phosphatases PP1 and Ssu72. PMID: 24945319
  2. Ubiquitination of SWD2 links H2B ubiquitination to H3K4 trimethylation. PMID: 18849979
Database Links

KEGG: sce:YKL018W

STRING: 4932.YKL018W

Protein Families
WD repeat SWD2 family
Subcellular Location
Nucleus. Chromosome, telomere.

Q&A

What is SWD2 and what are its primary functions in Saccharomyces cerevisiae?

SWD2 is an essential WD40 repeat protein in Saccharomyces cerevisiae that functions in two distinct protein complexes:

  • Set1/COMPASS (Complex of Proteins Associated with Set1) complex involved in histone H3 lysine 4 (H3K4) methylation

  • APT (Associated with Pta1) complex involved in RNA 3' end processing and termination

In the COMPASS complex, SWD2 contributes to efficient methylation of H3K4, particularly the di- and trimethylation states. SWD2 is unique among COMPASS components in that it is essential for viability, though this requirement stems primarily from its role in the APT complex rather than its COMPASS function . SWD2 plays a crucial role in mediating the interaction between the COMPASS complex and RNA polymerase II C-terminal domain (CTD), facilitating the recruitment of COMPASS to actively transcribed regions .

How does SWD2 contribute to the COMPASS complex structure and function?

SWD2 interacts with the N-terminal region of Set1 (specifically amino acids 124-229) in the COMPASS complex . This interaction is critical for multiple aspects of COMPASS function:

  • The Set1 N-terminal region and SWD2 together form a module that mediates interaction with the RNA polymerase II CTD

  • This interaction is crucial for proper recruitment of COMPASS to chromatin and subsequent H3K4 methylation

  • The WD40 domain of SWD2, particularly residues around phenylalanine 250, is important for maintaining COMPASS integrity

Point mutations in the SWD2 WD40 domain can disrupt its association with Set1 and impair the recruitment of COMPASS to chromatin . Protein interaction studies and cryo-electron microscopy have confirmed that the Swd2–Set1 N-terminal module may fold over the catalytic body of COMPASS to contact other subunits and more C-terminal Set1 regions .

What domains are present in SWD2 and how do they function?

SWD2 is a 37-kDa protein containing six WD repeats, which are domains of about 40 amino acids that usually end with tryptophan-aspartic acid (WD) . The structure and function of these domains include:

  • Structure: WD repeat proteins typically fold into circularized β-propeller-like structures

  • Interaction capability: These structures can interact sequentially or simultaneously with several different proteins

  • Critical regions: Phenylalanine 250, located at the center of the WD40 domain at the tip of propeller blade 6, is particularly important for COMPASS association

DomainLocationFunctionEffect of Mutation
WD40 domainFull protein structureMediates protein-protein interactionsF250A mutation reduces association with Set1
Ubiquitylation sitesLysines 68 and 69Regulates H3K4 trimethylationK68/69 mutations reduce H3K4me3
Set1 binding regionThroughout WD40 structureInteracts with Set1 N-terminal regionMutations disrupt COMPASS integrity

Mutations in the WD40 domain (e.g., F250A) reduce SWD2's association with COMPASS, diminish H3K4 methylation activity, and impair binding to RNA polymerase II CTD .

What are the optimal methods to express and purify recombinant SWD2 in yeast systems?

For successful expression and purification of recombinant SWD2 in Saccharomyces cerevisiae, the following protocol is recommended:

Expression System Setup:

  • Subclone the SWD2 gene into a galactose-inducible expression vector like pYeastPro (approximately 8.7kb)

  • Transform the vector into a suitable yeast strain (e.g., YMY1032) using standard lithium acetate transformation protocols

  • Heat-shock the transformation mixture at 42°C for 15-30 minutes to increase transformation efficiency

  • Select transformants on SMD DO plates lacking uracil and incubate at 30°C for 48 hours

Protein Expression Protocol:

  • For small-scale testing, grow cultures in selection media with 2% raffinose

  • Induce protein expression by adding galactose to a final concentration of 2%

  • For large-scale production, transfer successful small-scale cultures to 100ml of 2% raffinose media

Purification Strategy:

  • Harvest cells by centrifugation after optimal induction time

  • Lyse cells using mechanical disruption in buffer containing protease inhibitors

  • Purify using affinity chromatography (if tagged) followed by size exclusion chromatography

  • For SWD2, which functions in protein complexes, consider co-expression with other COMPASS components to improve solubility and functionality

Optimization Notes:

  • For proteins with low yield, consider modifying with fusion tags or optimizing induction conditions

  • Monitor protein stability carefully, as SWD2 can be subject to degradation

  • For functional studies, consider purifying the entire COMPASS complex rather than SWD2 alone

How can researchers generate and characterize mutations in SWD2 to study its function?

To generate and analyze mutations in SWD2 for functional studies, researchers can employ several strategic approaches:

Mutation Generation Methods:

  • Site-directed mutagenesis:

    • Create specific point mutations like F250A in the WD40 domain, which affects SWD2's interaction with COMPASS

    • Target lysines 68 and 69, which are ubiquitylation sites crucial for H3K4 methylation

  • Domain manipulation:

    • Delete or replace specific domains to understand their function

    • Create chimeric proteins by swapping domains with homologs (e.g., using S. pombe SWD2 homologs which are complex-specific)

  • Genomic engineering:

    • Use CRISPR-Cas9 genome editing to introduce mutations at the endogenous locus

    • For the essential SWD2 gene, use plasmid shuffling where a wild-type copy is maintained on a URA3-marked plasmid while introducing mutants on another plasmid, followed by 5-FOA selection

Functional Characterization Approaches:

  • Protein interaction analysis:

    • Co-immunoprecipitation to test interactions with COMPASS components and RNA polymerase II

    • Yeast two-hybrid assays to map interaction domains

  • H3K4 methylation assessment:

    • Western blotting for global H3K4me levels

    • ChIP-qPCR for genomic distribution of methylation marks at specific loci like PMA1

    • In vitro histone methyltransferase assays with reconstituted complexes

  • Chromatin association:

    • ChIP to measure recruitment of mutant SWD2 and other COMPASS components to chromatin

    • Compare occupancy patterns with RNA polymerase II distribution

  • Cellular phenotypes:

    • Growth defects in different conditions

    • Transcriptional profiling to identify affected genes

    • For non-lethal mutations, assess meiotic phenotypes

Example Mutation Analysis:
The F250A mutation in SWD2's WD40 domain provides a valuable case study - while protein levels remain similar to wild-type, this mutation causes:

  • Reduced H3K4me2/me3 levels

  • Decreased association with COMPASS

  • Diminished interaction with RNA polymerase II CTD

  • Reduced enrichment of COMPASS components at gene 5' regions

What techniques are used to study the interaction between SWD2 and the RNA polymerase II CTD?

Several sophisticated techniques can be employed to investigate the interaction between SWD2 and the RNA polymerase II CTD:

1. Yeast Two-Hybrid (Y2H) Assays:

  • Test interactions between Gal4 AD-CTD fusion and Gal4 BD fused to Set1 or its fragments

  • Compare interactions in wild-type SWD2 versus swd2Δ backgrounds (the lethality of swd2Δ can be suppressed by expression of a fragment from the termination factor Sen1)

  • Quantify interaction strength using reporter gene activation (e.g., HIS3 reporter)

2. Co-Immunoprecipitation (Co-IP):

  • Immunoprecipitate SWD2 or COMPASS components and detect CTD association

  • Use phospho-specific antibodies to determine if the interaction depends on specific CTD phosphorylation states (e.g., Ser5P)

  • Perform reciprocal IPs with RNA polymerase II antibodies to detect SWD2/COMPASS components

  • Compare wild-type to mutant strains (e.g., F250A SWD2 mutant)

3. Domain-Specific Analysis:

  • Delete specific regions (e.g., Set1 1-200) and test effects on CTD binding

  • Create chimeric proteins by replacing the Set1 N-terminal region with the Nrd1 CTD-interacting domain (CID) to test functional complementation

  • Delete amino acids 200-210 from Set1 to specifically disrupt interaction with both SWD2 and the CTD

4. Genetic and Functional Analysis:

  • Test the independence of SWD2-mediated interactions from other factors

  • Compare RNApII co-precipitation with wild-type Set1, SΔ200, and NSΔ200 fusion proteins in SWD2 versus swd2Δ backgrounds

  • Analyze the functional consequences of disrupted interactions through H3K4 methylation assays

5. In Vitro Binding Studies:

  • Use purified recombinant proteins to test direct binding between SWD2, Set1 fragments, and synthetic CTD peptides with different phosphorylation states

  • Employ surface plasmon resonance or isothermal titration calorimetry to quantify binding affinities and kinetics

Key finding: The evidence suggests that the N-terminal region of Set1 and SWD2 cooperatively interact with RNApII-CTD to promote proper recruitment of COMPASS to transcription elongation complexes at 5' ends of genes .

How does SWD2 ubiquitylation affect COMPASS function and H3K4 methylation?

SWD2 ubiquitylation represents a critical regulatory mechanism that connects histone H2B ubiquitylation to H3K4 methylation:

Biochemical Mechanism:

  • SWD2 is ubiquitylated at lysine residues 68 and 69

  • This modification depends on prior histone H2B monoubiquitylation

  • The same Rad6/Bre1 ubiquitylation enzymes responsible for H2B ubiquitylation directly participate in SWD2 modification

Functional Consequences:
Preventing SWD2 ubiquitylation by mutating K68 and K69 does NOT affect:

  • Set1 stability

  • The interaction between SWD2 and Set1

  • The ability of SWD2 to interact with chromatin

  • H3K4 trimethylation at the 5' end of transcribing genes (major reduction)

  • H3K4 dimethylation (lesser reduction)

  • H3K4 monomethylation (unaffected)

Mechanistic Insight:
SWD2 ubiquitylation controls the recruitment of Spp1, a COMPASS subunit that is necessary for trimethylation . This creates a regulatory pathway where:

  • H2B is ubiquitylated by Rad6/Bre1 at active genes

  • This leads to SWD2 ubiquitylation by the same enzymes

  • Ubiquitylated SWD2 promotes Spp1 association with COMPASS

  • Spp1 enables COMPASS to perform H3K4 trimethylation

  • This establishes the characteristic pattern of H3K4me3 at 5' ends of active genes

SWD2 thus serves as a key factor in the trans-histone pathway connecting H2B ubiquitylation to H3K4 methylation, functioning as a major H3-binding component of COMPASS .

What is the molecular mechanism by which SWD2 and the Set1 N-terminal region recruit COMPASS to actively transcribed genes?

The recruitment of COMPASS to actively transcribed genes involves a sophisticated coordination between SWD2, the Set1 N-terminal region, and the RNA polymerase II machinery:

Key Components of the Recruitment Mechanism:

  • CTD Recognition Module:

    • The Set1 N-terminal region (amino acids 1-200) and SWD2 together form a module that mediates interaction with the Rpb1 CTD

    • This interaction preferentially occurs with Ser5-phosphorylated CTD, which is enriched at the 5' ends of transcribed genes

    • Deletion of the first 200 amino acids of Set1 results in both loss of RNApII CTD binding and reduction of H3K4me2/me3 levels

  • Cooperative Interaction:

    • Neither SWD2 nor the Set1 N-terminal region alone is sufficient for robust CTD binding

    • Yeast two-hybrid assays show that SWD2 deletion severely reduces, but does not abolish, the interaction between Set1 and the CTD

    • This suggests either a composite binding surface or that one component triggers a CTD-binding conformation in the other

  • Critical Domains:

    • Deleting amino acids 200-210 from Set1 weakens its interaction with both SWD2 and the CTD

    • The WD40 domain of SWD2, particularly residue F250, is important for proper Set1 interaction and subsequent CTD binding

Experimental Evidence:

Experimental ApproachKey FindingReference
Yeast two-hybridSet1 N-terminal region (F1 fragment) interacts with CTD
Co-immunoprecipitationSWD2 deletion reduces Set1-CTD interaction
CTD binding domain swapNrd1 CID can partially substitute for Set1 N-terminal region
Point mutation analysisSWD2 F250A reduces Set1-CTD interaction
ChIP-qPCRSWD2 F250A diminishes COMPASS recruitment to 5' gene regions

Functional Validation:
The Nrd1 CID fusion experiment provides compelling evidence for this model. Replacing the Set1 N-terminal region with the Nrd1 CTD-interacting domain (NSΔ200) partially restores H3K4 methylation and CTD binding. Importantly, this fusion protein interacts with RNApII independently of SWD2, confirming that CTD binding is the primary function of the Set1-SWD2 module .

This recruitment mechanism represents the initial step in COMPASS function, which is followed by activation of its methyltransferase activity through the H2B ubiquitylation-dependent pathway.

What is the role of SWD2 in meiosis compared to its function in vegetative cells?

The role of SWD2 in meiosis versus vegetative growth reveals interesting distinctions in COMPASS complex requirements across different cellular processes:

SWD2 in Vegetative Cells:

  • Complex Membership:

    • Essential component of both COMPASS (histone methylation) and APT (RNA processing) complexes

    • In COMPASS, interacts with the Set1 N-terminal region (amino acids 124-229)

  • H3K4 Methylation Role:

    • Required for H3K4 di- and trimethylation

    • Depletion strongly destabilizes Set1 and reduces H3K4 methylation

    • Contributes to COMPASS recruitment via RNA polymerase II CTD interaction

  • Gene Regulation:

    • Contributes to position-dependent gene silencing at telomeres, mating type locus, and ribosomal DNA

    • Helps establish H3K4 methylation patterns in active coding regions

SWD2 in Meiosis:

  • Differential COMPASS Requirements:

    • COMPASS subunits show distinct requirements during meiotic progression

    • Core COMPASS members (Set1, Swd1, Swd3) have specialized meiotic functions

  • Temporal Specialization:

    • Set1 and Swd1 are required for progression through early meiosis (prophase I and chromosome segregation)

    • Swd3 is critical for late meiosis and spore morphogenesis

    • By association, SWD2 may have stage-specific roles in the meiotic program

  • Methylation-Independent Functions:

    • The meiotic requirement for Set1 appears to be independent of H3K4 methylation, suggesting non-histone substrates or structural roles

    • This may extend to SWD2's function within the COMPASS complex during meiosis

  • Complex Remodeling:

    • Evidence suggests COMPASS undergoes remodeling during stress response and meiosis

    • This may involve altered roles or interactions of SWD2 during meiotic progression

Research Implications:
The distinct requirements for COMPASS subunits during meiosis suggest that SWD2 may have specialized functions in this process compared to vegetative growth. Future research should specifically investigate SWD2's contribution to meiotic progression, potentially through the creation of meiosis-specific conditional alleles or degradation systems that allow temporal control of SWD2 activity during different meiotic stages.

What are the common challenges in studying SWD2 function and how can they be overcome?

Investigating SWD2 function presents several technical challenges that require strategic experimental approaches:

Challenge 1: Essential Gene Status

  • Problem: SWD2 is essential for viability in yeast, making it difficult to create complete knockout strains.

  • Solutions:

    • Use conditional alleles or temperature-sensitive mutants

    • Employ auxin-inducible degron systems for controlled depletion

    • Express a fragment from the termination factor Sen1 (amino acids 1890-2092) to suppress swd2Δ lethality

    • Create point mutations that affect specific functions while maintaining viability

Challenge 2: Dual Complex Membership

  • Problem: SWD2 functions in both COMPASS and APT complexes, complicating phenotype interpretation.

  • Solutions:

    • Create specific mutations that selectively disrupt one interaction but not the other

    • Use the F250A mutation in the WD40 domain that primarily affects COMPASS association

    • Study S. pombe homologs, which are complex-specific, to inform S. cerevisiae research

    • Employ complex-specific immunoprecipitation to analyze differential association

Challenge 3: Protein Stability Issues

  • Problem: Mutations in SWD2 or its interacting partners can lead to protein degradation.

  • Solutions:

    • Monitor protein levels by Western blotting

    • Use proteasome inhibitors when appropriate

    • For Set1, whose stability depends on SWD2, use N-terminal truncations (e.g., SΔ200) that remain stable even in swd2Δ cells

    • Add stabilizing protein fusion tags

Challenge 4: Reconstituting Functional Complexes

  • Problem: Recombinant expression may yield non-functional protein.

  • Solutions:

    • Co-express SWD2 with other COMPASS components

    • Use the galactose induction system in S. cerevisiae for proper expression

    • Optimize induction conditions and expression time

    • Consider testing protein functionality via in vitro methyltransferase assays

Challenge 5: Post-translational Modifications

  • Problem: SWD2 function is regulated by ubiquitylation, which can be difficult to preserve during purification.

  • Solutions:

    • Develop in vitro ubiquitylation systems with Rad6/Bre1 enzymes

    • Create lysine mutants (K68R/K69R) to study the effects of preventing ubiquitylation

    • Use deubiquitylase inhibitors during protein extraction

Challenge 6: Technical Challenges in Interaction Studies

  • Problem: Some interactions, like with RNA polymerase II CTD, may be difficult to detect.

  • Solutions:

    • Combine multiple approaches (Y2H, Co-IP, in vitro binding)

    • Use domain swap experiments, such as the Nrd1 CID fusion to Set1Δ200

    • Perform crosslinking before immunoprecipitation to capture transient interactions

How can researchers differentiate between the roles of SWD2 in COMPASS versus its function in the APT complex?

Distinguishing between SWD2's functions in COMPASS versus the APT complex requires carefully designed experimental strategies:

Genetic Approaches:

  • Separation of function alleles:

    • Generate and screen for SWD2 mutations that selectively impair one function while preserving the other

    • The F250A mutation in the WD40 domain primarily disrupts COMPASS association while maintaining viability

    • Create a library of point mutations throughout SWD2 and assess their differential effects on COMPASS and APT functions

  • Bypass strategies:

    • The lethality of swd2Δ can be suppressed by expressing a fragment from the termination factor Sen1 (amino acids 1890-2092)

    • This allows study of COMPASS functions in the absence of SWD2

    • For COMPASS bypass, the NSΔ200 fusion (Nrd1 CID fused to Set1Δ200) enables CTD interaction independent of SWD2

Biochemical Methods:

  • Complex-specific immunoprecipitation:

    • Use antibodies against COMPASS-specific components (e.g., Set1, Spp1) or APT-specific factors

    • Compare wild-type and mutant SWD2 association with each complex

    • Analyze post-translational modifications of SWD2 in each complex

  • Function-specific readouts:

    ComplexFunctional ReadoutMethodology
    COMPASSH3K4 methylationWestern blot, ChIP-qPCR/seq
    COMPASSSet1 stabilityWestern blot, protein half-life
    APTRNA 3' processing3' RACE, RNA-seq
    APTTranscription terminationNET-seq, TT-seq
  • Structural analysis:

    • Define the binding interfaces for SWD2 in each complex

    • Use crosslinking mass spectrometry to identify residues involved in specific interactions

    • Apply this information to design complex-specific mutations

Advanced Approaches:

  • Evolutionary insights:

    • Study S. pombe, which has two SWD2 homologs (one for each complex)

    • Apply knowledge of the complex-specific homologs to identify critical residues in S. cerevisiae SWD2

  • Temporal regulation:

    • Use rapid depletion systems to determine which SWD2 functions are affected first

    • This might reveal direct versus indirect roles in each complex

  • Genomic approaches:

    • Compare genome-wide maps of SWD2 occupancy with COMPASS-specific (H3K4me3) and APT-specific markers

    • Identify sites where SWD2 functions in one complex but not the other

    • Analyze differential gene expression upon complex-specific mutations

What is known about the structural basis for SWD2 interactions with Set1 and other COMPASS components?

The structural basis for SWD2 interactions with Set1 and other COMPASS components reveals sophisticated molecular mechanisms underlying complex assembly and function:

SWD2-Set1 Interaction Architecture:

  • Binding Regions:

    • SWD2 interacts primarily with the N-terminal region of Set1, specifically amino acids 124-229

    • Deleting amino acids 200-210 from Set1 weakens its interaction with SWD2

    • The WD40 domain of SWD2, particularly around phenylalanine 250, is critical for Set1 interaction

  • Structural Organization:

    • Low-resolution cryo-EM structural analysis confirms that the Swd2–Set1 N-terminal module folds over the catalytic body of COMPASS

    • This module contacts other subunits and more C-terminal Set1 regions

    • The propeller-like structure of the WD40 domains in SWD2 likely creates multiple protein interaction surfaces

  • Functional Domains:

    • The F250A mutation in SWD2's WD40 domain strongly reduces its association with COMPASS

    • Reconstituted recombinant Set1/COMPASS complex incorporates lower levels of F250A than wild-type SWD2

    • This mutation also impairs H3K4 methyltransferase activity in vitro

Complex Assembly Dynamics:

Structural-Functional Hypotheses:

  • Allosteric Regulation Model:

    • The positioning of the Swd2–Set1 N-terminal module may regulate COMPASS activity

    • CTD binding could move this module, potentially relieving autoinhibition by making the Set1 active site more accessible

    • This would connect transcription (via CTD binding) directly to methyltransferase activation

  • H3 Binding Interface:

    • SWD2 appears to be a major H3-binding component of COMPASS

    • This positions it to sense chromatin states and contribute to substrate recognition

    • The WD40 structure may create a platform for histone tail binding

These structural insights provide a foundation for understanding COMPASS assembly, recruitment, and regulation, though high-resolution structures of the complete complex would further clarify these mechanisms.

How can SWD2 research contribute to understanding broader epigenetic mechanisms in eukaryotes?

Research on SWD2 provides critical insights into fundamental epigenetic regulatory mechanisms that are conserved across eukaryotes:

Trans-Histone Modification Pathways:

  • H2B-H3K4 Crosstalk:

    • SWD2 research revealed a key molecular mechanism linking H2B ubiquitylation to H3K4 methylation

    • SWD2 ubiquitylation at K68/K69 provides the missing link in this trans-histone pathway

    • This pathway represents a paradigm for how modifications on one histone can influence modifications on another

  • Regulatory Networks:

    • The Rad6/Bre1 ubiquitylation machinery targets both H2B and SWD2

    • This creates a coordinated regulatory network connecting different epigenetic modifications

    • Understanding SWD2's role helps map how these networks are integrated

Transcription-Coupled Epigenetic Regulation:

  • Connecting Transcription to Chromatin:

    • SWD2 and Set1 N-terminal region form a module that interacts with the RNA polymerase II CTD

    • This mechanism explains how transcription directly influences chromatin modification

    • The preferential binding to Ser5P-CTD explains the enrichment of H3K4me3 at 5' ends of genes

  • Temporal Coordination:

    • SWD2's dual role in COMPASS and RNA processing/termination suggests coordination between these processes

    • This supports models where chromatin modification, transcription, and RNA processing are temporally and spatially regulated as a unit

Evolutionary Conservation and Specialization:

  • Conserved Mechanisms:

    • The SWD2 homolog Wdr82 in mammals performs similar functions in Set1/COMPASS recruitment

    • This conservation underscores the fundamental importance of these pathways

    • Research in yeast SWD2 directly informs understanding of mammalian Wdr82 function

  • Evolutionary Specialization:

    • S. pombe has evolved two separate SWD2 homologs for COMPASS and APT functions

    • This specialization reveals how epigenetic regulation can evolve and adapt

    • Comparing species with one versus two SWD2 homologs provides insights into complex evolution

Developmental and Cell-Type Specific Regulation:

  • Meiosis-Specific Functions:

    • COMPASS components show distinct requirements in meiosis versus mitosis

    • This suggests SWD2 may have specialized functions in different cellular contexts

    • Understanding these differences will illuminate how epigenetic regulation is adapted to specific biological processes

  • Beyond Histone Methylation:

    • The meiotic requirement for Set1 appears independent of H3K4 methylation

    • This suggests non-histone substrates or structural roles for COMPASS

    • SWD2 research may reveal new targets and functions beyond traditional histone modification

By elucidating these mechanisms, SWD2 research contributes to our fundamental understanding of epigenetic regulation across eukaryotes, with implications for development, cell differentiation, and disease.

What are the unresolved questions regarding SWD2 function that require further investigation?

Despite significant advances in understanding SWD2, several important questions remain unresolved and warrant further investigation:

Structural Unknowns:

  • High-resolution structure of SWD2 in COMPASS:

    • How exactly does SWD2 position itself within the complete COMPASS complex?

    • What is the precise molecular interface between SWD2 and Set1?

    • How does SWD2 ubiquitylation structurally affect Spp1 recruitment?

  • CTD binding mechanism:

    • What is the exact structural basis for the cooperative binding of Set1-SWD2 to the RNA polymerase II CTD?

    • Does this involve a composite binding surface or allosteric regulation?

    • Which specific residues in SWD2 and Set1 mediate this interaction?

Functional Mysteries:

  • Dual complex regulation:

    • How is SWD2 partitioned between COMPASS and APT complexes?

    • Is there competition between these complexes for limited SWD2?

    • Do post-translational modifications regulate SWD2's distribution between complexes?

  • Meiotic functions:

    • What are the specific roles of SWD2 during meiotic progression?

    • Does SWD2 contribute to the methylation-independent functions of Set1 during meiosis?

    • How is SWD2 function modified during meiosis compared to mitotic growth?

  • Non-histone substrates:

    • Does COMPASS, with SWD2, have non-histone methylation targets?

    • Could SWD2 help target Set1 to these alternative substrates?

Regulatory Pathways:

  • Beyond ubiquitylation:

    • What other post-translational modifications regulate SWD2?

    • How do these modifications cross-talk with SWD2 ubiquitylation?

    • Are there additional readers or writers that interact with modified SWD2?

  • Cell cycle regulation:

    • How is SWD2 function regulated through the cell cycle?

    • Does SWD2 contribute to cell cycle-dependent changes in H3K4 methylation patterns?

  • Stress response:

    • How is SWD2 function affected during stress conditions?

    • Does SWD2 contribute to COMPASS remodeling during stress response?

Evolutionary Questions:

  • SWD2 homolog specialization:

    • What evolutionary pressures led to the specialization of SWD2 homologs in some species?

    • What are the functional consequences of having dedicated homologs versus a single protein with dual functions?

  • Conservation of regulatory mechanisms:

    • Are the ubiquitylation-dependent regulatory mechanisms for SWD2 conserved in higher eukaryotes?

    • Does human Wdr82 function through similar mechanisms?

Technical Challenges:

Addressing these questions will require innovative approaches combining structural biology, biochemistry, genetics, and genomics to fully understand SWD2's multifaceted roles in epigenetic regulation.

How can studying recombinant SWD2 contribute to understanding gene amplification mechanisms in Saccharomyces cerevisiae?

Studying recombinant SWD2 provides unique insights into gene amplification mechanisms in Saccharomyces cerevisiae, with broader implications for genome dynamics and adaptation:

SWD2 as a Model for Gene Amplification Studies:

  • Amplification in Experimental Evolution:

    • Studies of recombinant genes in S. cerevisiae have revealed fascinating amplification mechanisms that may apply to SWD2 research

    • Heterologous genes (like XylA) inserted near ARS sequences can undergo ~9-fold amplification during adaptive evolution

    • Similar mechanisms could be applied to study SWD2 function through controlled gene dosage

  • Mechanisms of Gene Amplification:

    • Formation of eccDNA (extrachromosomal circular DNA elements) has been observed even without repetitive sequences

    • These elements can use micro-homology sequences as short as 8 nucleotides

    • Such mechanisms could be leveraged to create strains with variable SWD2 copy numbers for functional studies

Practical Applications for SWD2 Research:

  • Controlled Expression Systems:

    • The galactose induction system used for recombinant protein expression can be optimized for SWD2

    • Gene amplification could provide enhanced expression for biochemical and structural studies

    • Multiple integrated copies could overcome limitations of single-copy expression

  • Stability and Plasmid Design:

    • Studies on plasmid stability in S. cerevisiae inform optimal systems for SWD2 expression

    • Semi-defined media with controlled yeast extract concentrations can maintain high plasmid stability

    • Mathematical models can predict biomass concentration, protein expression, and plasmid stability

Technical Approaches:

  • Detecting Gene Amplification:

    • PCR methods using outward-facing primers to detect circular or tandem repeat formation

    • Southern blot analysis to confirm copy number variations

    • Next-generation sequencing to map precise amplification boundaries

  • Creating Amplification Systems:

    • Strategic placement of ARS elements near SWD2 to promote amplification

    • Selection strategies to favor cells with amplified SWD2

    • Temporal analysis of gene amplification during adaptation experiments

Research Questions Addressable Through Gene Amplification:

  • Dosage Effects:

    • How does increased SWD2 dosage affect the balance between COMPASS and APT complex formation?

    • Is there a dosage threshold where SWD2 functions become saturated?

    • Does overexpression of SWD2 alter global H3K4 methylation patterns?

  • Adaptation Mechanisms:

    • Could cells adapt to challenges in epigenetic regulation through SWD2 amplification?

    • How might gene amplification mechanisms contribute to evolutionary responses involving chromatin regulation?

  • Complex Assembly Dynamics:

    • Does increased SWD2 availability alter the stoichiometry of COMPASS or APT complexes?

    • Could this approach reveal rate-limiting steps in complex assembly?

By leveraging the natural gene amplification mechanisms of S. cerevisiae, researchers can develop novel approaches to studying SWD2 function, potentially revealing new aspects of its role in chromatin regulation and RNA processing.

What are the key takeaways from current research on recombinant Saccharomyces cerevisiae COMPASS component SWD2?

Current research on the COMPASS component SWD2 in Saccharomyces cerevisiae has yielded several significant insights that advance our understanding of epigenetic regulation:

Structural and Functional Insights:

  • SWD2 is a WD40 repeat protein that functions in two distinct complexes: COMPASS (histone methylation) and APT (RNA processing), with essential viability stemming from its APT role .

  • In COMPASS, SWD2 interacts with the N-terminal region of Set1 (amino acids 124-229) to form a module that mediates interaction with the RNA polymerase II CTD, particularly when phosphorylated at serine 5 .

  • The WD40 domain of SWD2, especially residue F250, is critical for maintaining COMPASS integrity and proper recruitment to chromatin .

  • SWD2 is ubiquitylated at lysines 68 and 69 by the same Rad6/Bre1 enzymes that ubiquitylate H2B, creating a direct link in the trans-histone pathway connecting H2B ubiquitylation to H3K4 methylation .

  • This ubiquitylation of SWD2 controls the recruitment of Spp1, a COMPASS subunit necessary for H3K4 trimethylation, without affecting SWD2's interaction with Set1 or chromatin .

Mechanistic Discoveries:

  • The Set1 N-terminal region and SWD2 cooperatively interact with the RNA polymerase II CTD, as neither component alone shows robust binding .

  • Replacing the Set1 N-terminal region with the Nrd1 CTD-interacting domain partially restores COMPASS recruitment and H3K4 methylation, indicating that CTD binding is a major function of this region .

  • Even when COMPASS is recruited via the Nrd1 CID bypass mechanism, H2B ubiquitylation is still required for efficient H3K4 methylation, demonstrating that H2Bub acts downstream of initial COMPASS recruitment .

  • COMPASS subunits, including SWD2, show distinct requirements during meiosis compared to vegetative growth, with evidence that Set1 has methylation-independent functions in meiosis .

Technical Advancements:

  • Expression systems for recombinant proteins in S. cerevisiae using galactose induction provide effective methods for studying SWD2 and other COMPASS components .

  • Point mutations like F250A in SWD2's WD40 domain provide valuable tools for disrupting specific functions while maintaining protein expression .

  • Plasmid stability studies inform optimal conditions for maintaining recombinant SWD2 expression systems .

  • Gene amplification mechanisms in S. cerevisiae, involving eccDNA formation even without repetitive sequences, offer potential approaches for enhanced SWD2 expression and functional studies .

These findings establish SWD2 as a critical component in epigenetic regulation, connecting transcription, histone modification, and RNA processing through its dual complex membership and regulated interactions.

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.