The Recombinant Xenopus laevis Leucine-rich Repeat and WD Repeat-containing Protein 1 (lrwd1), partial is a recombinant protein derived from the African clawed frog, Xenopus laevis. This protein is a partial form of the full-length leucine-rich repeat and WD repeat-containing protein 1 (LRWD1), which plays a crucial role in chromatin organization and DNA replication. LRWD1 is known to interact with the origin recognition complex (ORC) and is involved in heterochromatin silencing and the regulation of DNA replication origins.
LRWD1 contains two main structural domains: leucine-rich repeats (LRRs) and a WD40 repeat domain. The WD40 domain is crucial for the protein's interaction with histone marks, such as trimethylated lysine 9 of histone H3 (H3K9me3) and trimethylated lysine 20 of histone H4 (H4K20me3) . These interactions are essential for the recruitment of LRWD1 to heterochromatin regions, where it contributes to gene silencing and the maintenance of chromatin structure.
LRWD1 is associated with the origin recognition complex (ORC), which is involved in the initiation of DNA replication. The dynamic binding of LRWD1 to chromatin during the cell cycle, particularly during the G1 phase, suggests its role in regulating replication origins . The protein's ability to bind repressive histone marks helps in the silencing of heterochromatic regions, ensuring proper chromatin organization and preventing inappropriate gene expression.
Binding Specificity: LRWD1 preferentially binds to trimethylated repressive histone marks, such as H3K9me3 and H4K20me3, in a manner dependent on its WD40 domain but independent of ORC proteins .
Chromatin Localization: The localization of LRWD1 to pericentric heterochromatin is dependent on H3K9me3 but not H4K20me3. Depletion of LRWD1 leads to increased transcription of major satellite repeats, highlighting its role in heterochromatin silencing .
The binding of LRWD1 to chromatin is temporally regulated during the G1 phase, with its protein levels peaking at early G1 and decreasing at the G1/S boundary .
This temporal regulation suggests that LRWD1 plays a role in the dynamic selection and licensing of replication origins.
The recombinant Xenopus laevis LRWD1 protein is produced with a purity of ≥85% . This partial form of the protein is used in research to study its interactions with chromatin and other proteins involved in DNA replication and chromatin organization.
| Feature | Description |
|---|---|
| Source | Xenopus laevis |
| Purity | ≥85% |
| Function | Chromatin organization, DNA replication regulation |
| Binding Specificity | H3K9me3, H4K20me3 |
| Role | Mechanism |
|---|---|
| Heterochromatin Silencing | Binds to H3K9me3, maintaining heterochromatin structure |
| DNA Replication Regulation | Interacts with ORC, influencing replication origin selection |
KEGG: xla:444544
UniGene: Xl.43573
Xenopus laevis LRWD1 is a 722 amino acid protein with a molecular mass of approximately 82.1 kDa . The protein contains characteristic leucine-rich repeats (LRRs) and WD40 repeat domains that facilitate protein-protein interactions and chromatin binding. The WD40 domain specifically enables interaction with repressive histone marks and serves as a structural platform for assembling protein complexes . The full amino acid sequence is available in protein databases and begins with MTKLTTELLLKKGLPKCSHLKDLKK .
LRWD1 serves multiple critical cellular functions:
Required for G1/S transition in the cell cycle
Recruits and stabilizes the origin recognition complex (ORC) onto chromatin during G1 phase
Establishes pre-replication complex (preRC) at replication origins
Binds a combination of DNA and histone methylation repressive marks on heterochromatin
Required for silencing of major satellite repeats
Maintains heterochromatin regions through interactions with repressive marks
LRWD1 binding sites on chromatin are dynamic and temporally regulated during the G1 phase. Research indicates that LRWD1 association with specific genomic sites decreases as cells progress toward S-phase. Notably, the majority of LRWD1-bound sites represent replication origins that associate with repressive chromatin marks like H3K9me3 and methylated-CpGs, consistent with LRWD1-bound origins initiating DNA replication late in S-phase .
Expression of recombinant Xenopus laevis LRWD1 can be achieved through several systems:
Bacterial expression systems: For partial protein domains or shorter constructs
Use E. coli BL21(DE3) strains with pET-based vectors
Expression at lower temperatures (16-18°C) often improves solubility
Addition of fusion tags (His, GST, MBP) may enhance solubility and facilitate purification
Insect cell expression systems: For full-length functional protein
Baculovirus expression systems provide eukaryotic post-translational modifications
Sf9 or High Five cells are commonly used
Consider using a secretion signal for improved yield
Mammalian expression systems: For studies requiring native-like modifications
HEK293 or CHO cells with strong promoters (CMV)
Transient transfection for quick analysis or stable cell lines for consistent production
Each method requires optimization of expression conditions including temperature, induction time, and inducer concentration to maximize yield and maintain protein functionality .
Several complementary approaches can be employed to investigate LRWD1-chromatin interactions:
Chromatin Immunoprecipitation (ChIP):
Use specific anti-LRWD1 antibodies to pull down protein-DNA complexes
Combine with next-generation sequencing (ChIP-seq) to map genome-wide binding sites
Sequential ChIP can be used to identify co-occupancy with other proteins or histone marks
Proximity Ligation Assays (PLA):
Detect protein-protein interactions at specific genomic loci
Particularly useful for studying LRWD1 interaction with ORC components
In vitro binding assays:
Electrophoretic mobility shift assays (EMSA) with recombinant LRWD1
Surface plasmon resonance (SPR) to measure binding kinetics
Pull-down assays using synthetic oligonucleotides with specific modifications
Microscopy approaches:
LRWD1 functions as a critical coordinator between DNA replication and epigenetic regulation. Research demonstrates that LRWD1:
Directly associates with repressive histone marks, particularly H3K9me3, through its WD40 domain
Physically interacts with the enzymes that catalyze these repressive marks
Contributes to H3K9 methylation at specific genomic loci, as H3K9 methylation is diminished at LRWD1-H3K9me3 overlapping regions in LRWD1-depleted cells
Influences DNA methylation patterns, as altered DNA methylation is observed at LRWD1-occupied sites in cells lacking LRWD1
Is itself influenced by repressive chromatin marks that affect its binding to chromatin
This suggests a reciprocal relationship where LRWD1 helps establish heterochromatin while heterochromatin marks stabilize LRWD1 binding. This creates a reinforcing loop that maintains repressive chromatin environments at specific genomic loci, particularly late-firing replication origins .
Studies using knockout models provide insights into LRWD1's developmental roles:
Developmental expression: LRWD1 is ubiquitously expressed throughout most tissues during mouse embryonic development
Embryonic development: Surprisingly, LRWD1 depletion does not significantly impact embryonic development
Postnatal growth: Homozygous LRWD1 mutants display retarded postnatal growth compared to wild-type counterparts
Cellular proliferation: Mouse embryonic fibroblasts (MEFs) depleted of LRWD1 show reduced proliferation rates compared to wild-type cells
Gene expression effects:
LRWD1 knockout increases expression of epigenetically silenced repetitive elements
Minimal effect observed on protein-coding gene expression
These findings suggest LRWD1 plays an important but not essential role in postnatal development, likely through modulating DNA replication and maintaining epigenetic silencing of repetitive elements .
| Feature | Xenopus laevis LRWD1 | Mammalian LRWD1 (ORCA) |
|---|---|---|
| Length | 722 amino acids | Varies by species (~647 in humans) |
| Molecular weight | 82.1 kDa | ~70-75 kDa in mammals |
| Key domains | LRR and WD40 repeats | LRR and WD40 repeats |
| Primary function | G1/S transition, ORC stabilization | G1/S transition, ORC stabilization |
| Developmental role | Not fully characterized in Xenopus | Non-essential for embryonic development but important for postnatal growth in mice |
| Epigenetic function | Binds repressive marks | Binds repressive marks and coordinates heterochromatin formation |
| Cell cycle regulation | Associates with replication origins | Temporally regulated binding to late-firing origins |
Several complementary approaches can be used to identify and characterize LRWD1 binding sites:
ChIP-seq analysis pipeline:
Crosslink protein-DNA complexes with formaldehyde
Sonicate chromatin to 200-500bp fragments
Immunoprecipitate with anti-LRWD1 antibodies
Prepare libraries for next-generation sequencing
Bioinformatic analysis to identify enriched regions
Motif discovery to identify binding sequences
CUT&RUN or CUT&Tag alternatives:
Offers higher signal-to-noise ratio than traditional ChIP
Requires fewer cells
Provides higher resolution of binding sites
Integration with epigenomic data:
Overlay LRWD1 binding sites with histone modification data (especially H3K9me3)
Correlate with DNA methylation profiles
Compare binding patterns at different cell cycle stages
Validation approaches:
| Approach | Advantages | Limitations | Validation Methods |
|---|---|---|---|
| CRISPR/Cas9 knockout | Complete protein elimination; stable modification | Potential compensatory mechanisms; lethal if essential | Western blot; genomic PCR and sequencing; off-target analysis |
| siRNA/shRNA knockdown | Rapid implementation; titratable reduction | Transient effect; incomplete knockdown; off-target effects | Western blot; qRT-PCR; rescue experiments |
| Gene-trap insertion | Creates null alleles; useful for developmental studies | Limited targeting flexibility; potential hypomorphic effects | LacZ reporter expression; RT-PCR; Western blot |
| Conditional knockout | Tissue-specific or temporally controlled deletion | Complex breeding schemes; potential leakiness | Tissue-specific PCR; Western blot; immunohistochemistry |
| Degron systems | Rapid protein depletion; temporal control | Requires protein tagging; potential tag interference | Time-course Western blot; functional rescue |
When generating knockout models, researchers should:
Design multiple targeting strategies (e.g., different gRNAs for CRISPR)
Include proper controls (scrambled sequences, wild-type cells)
Validate knockout at DNA, RNA and protein levels
Perform rescue experiments with wild-type protein to confirm specificity
The observation that LRWD1 knockout increases expression of epigenetically silenced repetitive elements requires careful interpretation:
Direct vs. indirect effects:
Determine whether derepression is directly due to LRWD1 absence or secondary to other changes
Compare timing of LRWD1 depletion with onset of repetitive element expression
Examine co-localization of LRWD1 with affected repetitive elements
Element-specific analysis:
Not all repetitive elements respond equally to LRWD1 depletion
Categorize elements by type (LINEs, SINEs, LTRs, satellite repeats)
Quantify fold-changes for specific element families
Correlate with chromatin states at these loci
Functional consequences:
Evaluate genomic instability markers
Assess impact on nearby gene expression
Examine changes in nuclear organization
Consider potential activation of cellular stress responses
Methodological considerations:
To effectively capture LRWD1's dynamic behavior throughout the cell cycle:
Cell synchronization strategies:
Double thymidine block for G1/S boundary
Nocodazole treatment for M-phase
Serum starvation-release for G0/G1
Minimize synchronization artifacts by comparing multiple methods
Live-cell imaging approaches:
Generate fluorescently tagged LRWD1 (ensuring functionality is preserved)
Perform time-lapse microscopy through cell cycle
Combine with fluorescent cell cycle indicators
Quantify protein levels, localization, and mobility
Time-resolved ChIP-seq:
Perform ChIP-seq at defined time points after synchronization
Use spike-in controls for quantitative comparisons between time points
Generate genomic binding profiles at each cell cycle stage
Identify sites of dynamic vs. stable binding
Integration with replication timing data:
Despite significant progress in understanding LRWD1, several important questions warrant further investigation:
Structural biology:
Complete structural characterization of LRWD1 domains and their interactions
Structural basis for recognition of specific histone marks
Conformational changes during cell cycle progression
Regulatory mechanisms:
Post-translational modifications affecting LRWD1 function
Protein-protein interaction network throughout cell cycle
Mechanisms controlling LRWD1 expression and degradation
Evolutionary perspectives:
Functional conservation across species beyond mammalian models
Xenopus-specific adaptations in LRWD1 structure or function
Evolutionary relationship to other chromatin regulators
Disease relevance:
Emerging technologies offer promising approaches to address remaining questions:
Single-cell approaches:
Single-cell ChIP-seq or CUT&Tag for heterogeneity analysis
Single-cell multi-omics to correlate LRWD1 binding with transcription and chromatin state
Live-cell single-molecule tracking for binding dynamics
Proximity labeling methods:
BioID or TurboID fusions to map protein interaction network
Spatially-restricted enzymatic tagging to identify chromatin-bound partners
Integration with mass spectrometry for comprehensive interactome analysis
Genome engineering approaches:
CRISPR screens to identify genetic interactions
Base editing to introduce specific mutations in functional domains
Synthetic genomics to generate minimal systems for mechanistic studies
Computational approaches: