The Recombinant Danio rerio Chromodomain-helicase-DNA-binding protein 1-like (chd1l), partial refers to a recombinant form of the protein Chd1l derived from zebrafish (Danio rerio). Chd1l is a member of the chromatin remodeling family, specifically the Snf2-like family, and plays a crucial role in DNA repair and chromatin remodeling following DNA damage . This protein is unique due to its ability to bind poly(ADP-ribose) (PAR) through its macro domain, which is essential for its function in chromatin remodeling .
Chd1l contains a Snf2-like ATPase domain and a macro domain at its C-terminal end. The macro domain is responsible for binding PAR-modified proteins, which is critical for its chromatin remodeling activity . The protein's structure allows it to interact with damaged DNA sites, facilitating repair processes such as Base Excision Repair (BER) and Nucleotide Excision Repair (NER) .
ATPase Domain: Essential for chromatin remodeling.
Macro Domain: Binds PAR-modified proteins.
Role in DNA Repair: Facilitates chromatin relaxation and repair.
Chd1l is involved in several biological processes:
DNA Damage Response: Recruited to DNA damage sites to facilitate repair .
Chromatin Remodeling: Plays a role in chromatin relaxation, which is crucial for transcriptional regulation and DNA repair .
Developmental Processes: Implicated in early embryonic development, particularly in mouse embryo implantation .
Recent studies have highlighted the importance of Chd1l in various contexts:
Cancer: Overexpression of Chd1l is associated with tumor progression and poor prognosis in several cancers .
Neurodevelopment: Dosage imbalance of Chd1l contributes to neuroanatomical phenotypes, such as macrocephaly, and influences neuronal differentiation .
| Biological Process | Role of Chd1l | Implications |
|---|---|---|
| DNA Damage Response | Facilitates repair by chromatin remodeling | Essential for genomic stability |
| Chromatin Remodeling | Chromatin relaxation for transcriptional regulation | Influences gene expression and DNA repair |
| Developmental Processes | Involved in early embryonic development | Critical for proper embryonic development |
| Cancer | Associated with tumor progression and metastasis | Potential therapeutic target |
Recombinant forms of Chd1l, such as the partial protein from Danio rerio, are used in research to study its functions and interactions. These proteins can be used in biochemical assays to understand the mechanisms of chromatin remodeling and DNA repair. Additionally, they serve as tools for drug discovery, particularly in the development of inhibitors targeting Chd1l for cancer therapy .
CHD1L (Chromodomain helicase/ATPase DNA binding protein 1-like gene), also known as ALC1 (amplified in liver cancer 1) in humans, is a member of the SNF2-like subfamily of the SNF2 family of proteins . The full-length CHD1L protein contains four conserved domains:
SNF2_N domain: Involved in various cellular processes including DNA repair, chromatin unwinding, and transcription regulation
Helicase superfamily domain (HELICc): Contains the DEAD-like helicase superfamily ATP-binding region that participates in ATP-dependent DNA or RNA unwinding
Selenoprotein S (SelS) region: Found in many cell types and tissues
Macro domain: Recognizes poly ADP-ribose (PAR) which is crucial for its chromatin remodeling function
Unlike CHD1, CHD1L contains a macro domain that recognizes PAR rather than a chromo domain that recognizes methylated histone tails . This structural organization enables CHD1L to utilize energy from ATP hydrolysis to remodel chromatin, particularly at sites of DNA damage.
CHD1L plays a critical role in DNA repair through its PAR-dependent chromatin remodeling activity. The process follows these key steps:
Single-stranded DNA breaks (SSB) activate PARP1/2, which produces PAR at damage sites
CHD1L is recruited to these sites through its macro domain binding to PAR
Once recruited, CHD1L uses its ATPase activity to catalyze nucleosome sliding, leading to chromatin relaxation at damage sites
This chromatin relaxation facilitates access for DNA repair factors involved in Base Excision Repair (BER) and Nucleotide Excision Repair (NER)
CHD1L specifically mediates the efficient handover between PARP1/2, DNA glycosylases, and APEX1 downstream of lesion excision . Studies have shown that loss of CHD1L confers sensitivity to methyl-methanesulfonate, PARP inhibitors, and formyl-dU, and is synthetic lethal with homologous recombination deficiency (HRD) . This indicates CHD1L's essential role in maintaining genomic integrity through efficient DNA repair mechanisms.
Zebrafish models have revealed dose-dependent phenotypic effects of chd1l manipulation:
Overexpression effects: Overexpression of CHD1L in zebrafish embryos leads to macrocephaly (enlarged head) and increased larval body length
Deletion effects: Conversely, chd1l deletion results in microcephaly (smaller head) and decreased body length
These mirrored phenotypes have been consistently observed across zebrafish and mouse embryo models, confirming the conserved developmental function of CHD1L across vertebrates . The bidirectional, dose-dependent phenotypic effects highlight CHD1L's critical role in regulating head size and body growth during development, which correlates with the neuroanatomical and growth abnormalities observed in human 1q21.1 copy number variant carriers.
When working with recombinant Danio rerio CHD1L protein:
Storage considerations: Store working aliquots at 4°C and avoid repeated freeze-thaw cycles to maintain protein integrity and activity
ATP requirements: Ensure ATP availability in functional assays as CHD1L is an ATP-dependent chromatin remodeler
PAR dependency: Consider that CHD1L's helicase activity is strongly stimulated upon poly(ADP-ribose) binding, which may necessitate the inclusion of PARP1 and NAD+ in activity assays
Buffer composition: Use buffers that maintain protein stability while allowing for optimal enzymatic activity, typically including components that stabilize protein structure without interfering with the ATPase domain
Partial protein considerations: When working with partial recombinant protein, verify which domains are included and consider how this might affect functional assays compared to the full-length protein
Designing experiments to investigate CHD1L's role in neuronal differentiation requires a multifaceted approach:
CRISPR-Cas9 gene editing:
Neuronal differentiation assays:
Employ neural progenitor cell (NPC) differentiation protocols using wild-type and chd1l-modified cell lines
Monitor expression of neuronal markers by qPCR and immunostaining across differentiation timepoints
Assess morphological changes, neurite outgrowth, and synaptogenesis using high-content imaging
Transcriptomic analysis:
Perform RNA-seq at different stages of neuronal differentiation comparing control and chd1l-modified samples
Analyze differentially expressed genes (DEGs) using packages such as DESeq2, adjusting for batch effects
Focus on genes involved in neuronal differentiation and synaptogenesis
Use STRING analysis (string-db.org) and DECIPHER database (decipher.sanger.ac.uk/ddd/ddgenes) for gene annotation and pathway analysis
Chromatin accessibility studies:
In vivo developmental assessment:
Monitor telencephalon development in chd1l-modified zebrafish using lineage tracing and live imaging
Quantify head size and brain region measurements across developmental stages
Compare neurobehavioral outcomes in control versus chd1l-modified zebrafish
To analyze CHD1L's chromatin remodeling activity, researchers can employ these methodological approaches:
Nucleosome sliding assays:
Reconstitute positioned nucleosomes using purified histones and DNA containing specific positioning sequences
Incubate with recombinant CHD1L in the presence of ATP
Analyze nucleosome repositioning using native PAGE or restriction enzyme accessibility assays
Include controls with ATP-binding deficient CHD1L mutants
PARP1-dependent remodeling assays:
Set up in vitro systems containing CHD1L, PARP1, NAD+, and positioned nucleosomes
Induce DNA damage using UV or chemical agents to activate PARP1
Monitor chromatin relaxation and nucleosome repositioning in response to PAR synthesis
Measure ATPase activity in parallel to correlate with remodeling efficiency
Real-time fluorescence-based assays:
Employ FRET-labeled nucleosomes to monitor conformational changes during remodeling
Track remodeling kinetics in the presence and absence of PAR
Analyze the effect of PAR chain length and branching on CHD1L activation
Chromatin immunoprecipitation sequencing (ChIP-seq):
Map CHD1L binding sites genome-wide in control and DNA damage conditions
Correlate CHD1L occupancy with chromatin accessibility changes and repair factor recruitment
Analyze the relationship between CHD1L binding and transcriptional outcomes
Super-resolution microscopy:
Visualize CHD1L recruitment to DNA damage sites in live cells
Track the kinetics of chromatin relaxation following damage using fluorescently labeled histones
Correlate CHD1L activity with repair factor assembly at damage sites
CHD1L's interaction with transcriptional networks during zebrafish neurodevelopment can be characterized through these experimental approaches:
Integrated omics analysis:
Combine RNA-seq, ATAC-seq, and ChIP-seq data from chd1l-modified and control zebrafish embryos at key developmental stages
Identify transcription factors whose binding sites show altered accessibility in chd1l mutants
Use tools like LISA.cistrome for transcription regulator prediction based on differential gene expression patterns
Transcription factor binding studies:
CHD1L facilitates chromatin accessibility for pioneer transcription factors including SOX2 and OTX2 during forebrain regionalization
Simultaneously, it compacts chromatin through interaction with the repressor NuRD complex
This dual activity creates a precise balance of activation and repression required for proper telencephalon development
Gene Ontology enrichment analysis:
Apply PANTHER tool (geneontology.org) to identify biological processes enriched among CHD1L-regulated genes
Focus on neurodevelopmental pathways, particularly those involved in telencephalon development and neuronal differentiation
Visualize enrichment results using R package ggplot2 for clearer interpretation
Key target genes:
| Gene Category | Role in Neurodevelopment | CHD1L Regulatory Effect |
|---|---|---|
| Autism-associated genes | Synaptogenesis, neuronal connectivity | Direct regulation of chromatin accessibility |
| Telencephalon development genes | Forebrain regionalization | Enhanced accessibility for SOX2/OTX2 binding |
| Neuronal differentiation markers | Cell fate determination | Balanced activation/repression through NuRD interaction |
Temporal dynamics:
Monitor stage-specific effects of chd1l manipulation through developmental time series experiments
Track critical windows when CHD1L activity is essential for normal neuronal differentiation
Correlate molecular changes with the emergence of structural phenotypes like head size alterations
The relationship between CHD1L and poly(ADP-ribose) (PAR) in DNA repair involves several key molecular interactions:
Activation mechanism:
Recruitment dynamics:
DNA damage (particularly single-strand breaks) activates PARP1/2, which synthesizes PAR chains at damage sites
CHD1L is recruited to these sites through specific binding of its macrodomain to PAR
The temporal dynamics of this recruitment are rapid, making CHD1L one of the early responders to DNA damage
Functional consequences:
Once activated, CHD1L promotes chromatin relaxation at damage sites, creating accessibility for repair factors
CHD1L-dependent nucleosome remodeling facilitates the handover between PARP1/2, DNA glycosylases, and APEX1
This process is particularly important in Base Excision Repair (BER) and Nucleotide Excision Repair (NER)
Experimental verification:
The interaction can be verified through PAR-binding assays using recombinant CHD1L macrodomain
Mutations in the macrodomain that disrupt PAR binding (similar to those identified in CAKUT patients) demonstrate the importance of this interaction
The three variants identified in CAKUT patients (Gly700Arg, Ile765Met, and Ile827Val) all showed decreased interaction with PARP1 compared to wild-type CHD1L
Effective design and implementation of CHD1L knockout or knockdown studies in zebrafish requires careful consideration of these methodological aspects:
CRISPR-Cas9 knockout strategy:
Design multiple guide RNAs targeting early exons of the chd1l gene
Test guide RNA efficiency using in vitro cleavage assays
Inject Cas9 protein with validated guide RNAs into one-cell-stage zebrafish embryos
Screen F0 embryos for mutations using PCR and sequencing
Establish stable knockout lines through selective breeding of F0 founders
Morpholino-based knockdown approach:
Design splice-blocking or translation-blocking morpholinos specific to zebrafish chd1l
Determine optimal morpholino concentration through dose-response testing
Include appropriate controls (standard control morpholino and rescue experiments)
Validate knockdown efficiency through RT-PCR (for splice-blocking) or Western blot (for translation-blocking)
Phenotypic analysis pipeline:
Head size measurement: Use standardized imaging and analysis protocols to quantify head circumference at defined developmental stages
Body length assessment: Implement consistent methodology for measuring larval body length
Brain development: Apply neuroanatomical staining and imaging to assess specific brain regions, particularly the telencephalon
Behavioral testing: Conduct standardized assays for locomotor activity, response to stimuli, and learning behaviors
Molecular characterization:
Transcriptome analysis: Perform RNA-seq on whole embryos or isolated brain tissue from control and chd1l mutants
Chromatin accessibility: Implement ATAC-seq to identify regions with altered chromatin structure
Target gene validation: Use in situ hybridization and immunohistochemistry to confirm changes in expression of key neuronal and developmental genes
Rescue experiments:
Generate mRNA encoding wild-type human or zebrafish CHD1L
Co-inject with morpholinos or introduce into knockout backgrounds
Test domain-specific mutants (e.g., macro domain or ATPase domain mutants) to dissect functional requirements
Compare rescue efficiency with different CHD1L dosages to establish dose-dependency
Zebrafish CHD1L studies provide valuable insights into human 1q21.1 copy number variation (CNV) disorders through translational research approaches:
Phenotypic correlation:
The macrocephaly observed in CHD1L-overexpressing zebrafish mirrors the increased head circumference seen in humans with 1q21.1 duplications
Conversely, chd1l deletion in zebrafish causes microcephaly, similar to humans with 1q21.1 deletions
These mirrored phenotypes establish CHD1L as a critical dosage-sensitive gene within the 1q21.1 region
Transcriptional network analysis:
Transcriptomic studies in CHD1L-modified zebrafish and human cell models reveal disruption of genes involved in neuronal differentiation and synaptogenesis
Many affected genes overlap with known autism-associated genes, providing a molecular link between 1q21.1 CNVs and neurodevelopmental disorders
Gene Ontology analysis of these datasets highlights pathways relevant to the clinical manifestations of 1q21.1 CNV syndromes
Developmental mechanisms:
Zebrafish studies demonstrate that CHD1L regulates telencephalon development by modulating chromatin accessibility for key transcription factors like SOX2 and OTX2
This mechanism explains how CHD1L dosage affects brain size and potentially contributes to cognitive phenotypes in humans with 1q21.1 CNVs
The NuRD complex interaction suggests a balance between activation and repression that, when disrupted, leads to abnormal brain development
Therapeutic implications:
Understanding the downstream effects of CHD1L dosage imbalance identifies potential therapeutic targets
Zebrafish models allow for high-throughput screening of compounds that might mitigate the effects of CHD1L dosage abnormalities
Chromatin-modifying drugs could potentially normalize gene expression patterns disrupted by CHD1L imbalance
Studying CHD1L-PARP1 interactions during DNA repair presents several methodological challenges that require sophisticated experimental approaches:
Temporal resolution challenges:
The interaction occurs rapidly after DNA damage, requiring high temporal resolution techniques
Solutions include:
Synchronized damage induction using micro-irradiation coupled with live-cell imaging
Time-course sampling with rapid fixation to capture transient interactions
Development of biosensors that report CHD1L-PARP1 interactions in real-time
Spatial organization visualization:
Understanding the nanoscale organization of CHD1L and PARP1 at damage sites is challenging
Approaches to address this:
Super-resolution microscopy techniques like STORM or PALM
Proximity ligation assays to detect protein-protein interactions with spatial context
Correlative light and electron microscopy to combine functional and structural information
Biochemical reconstitution complexities:
Recapitulating the correct stoichiometry and activity of components in vitro is difficult
Strategies include:
Stepwise assembly of defined nucleosome substrates with purified proteins
Microfluidic approaches to control reaction conditions precisely
Use of nuclear extracts with defined genetic backgrounds (e.g., PARP1-null) to study dependency relationships
PAR chain heterogeneity:
PAR chains vary in length and branching, potentially affecting CHD1L recruitment and activation
Methods to address this:
Mass spectrometry to characterize PAR chain composition
Synthetic PAR chains with defined structures to test specificity
PAR-binding domain mutations to probe structural requirements for interaction
Genetic redundancy:
Functional overlap between CHD1L and other chromatin remodelers may mask phenotypes
Approaches to overcome this:
Combined depletion of multiple remodelers
Acute protein degradation systems like auxin-inducible degrons
Domain-specific mutations that affect specific functions while preserving others
CHD1L's influence on gene expression patterns during zebrafish embryogenesis encompasses multiple regulatory mechanisms:
Stage-specific transcriptional regulation:
CHD1L modulates chromatin accessibility at developmental gene promoters and enhancers
This creates permissive or restrictive chromatin states for stage-specific transcription factors
Experimental approaches should include:
Time-series RNA-seq and ATAC-seq throughout embryogenesis in wild-type and chd1l-modified embryos
In situ hybridization to spatially map expression changes of key developmental genes
Single-cell transcriptomics to identify cell type-specific effects
Developmental signaling pathway integration:
CHD1L likely coordinates responses to morphogenetic signals that drive embryonic patterning
Key pathways to investigate include:
Wnt signaling: Critical for anterior-posterior axis formation and telencephalon development
Sonic hedgehog (Shh): Important for ventral forebrain specification
Retinoic acid signaling: Essential for hindbrain patterning
Experiments should examine how CHD1L occupancy changes in response to pathway activation/inhibition
Epigenetic landscape shaping:
CHD1L participates in establishing developmental epigenetic patterns through:
CUT&RUN or CUT&Tag approaches can map CHD1L occupancy alongside histone modifications
Lineage-specific effects:
| Developmental Lineage | CHD1L Function | Experimental Approach |
|---|---|---|
| Neural ectoderm | Promotes telencephalon development | Neural-specific chd1l knockout |
| Neural crest | Influences migration and differentiation | Lineage tracing in chd1l mutants |
| Mesoderm | Affects somitogenesis and growth | Time-lapse imaging of somite formation |
| Endoderm | Potential role in organ development | Organ-specific marker analysis |
Differential sensitivity periods:
CHD1L function may be particularly critical during specific developmental windows
Conditional knockout or chemical inhibition approaches at different stages can identify these sensitive periods
Correlating molecular changes with morphological outcomes will reveal the developmental consequences of CHD1L dysregulation