SHPRH is a complex multidomain protein belonging to the RAD5/RAD16-related subgroup of SNF2 family proteins. Its primary function involves DNA repair through its E3 ubiquitin ligase activity, particularly in post-replication repair pathways. The protein contains several functional domains:
SNF2-family ATPase domain: Responsible for ATP hydrolysis
RING finger domain: Mediates E3 ubiquitin ligase activity
PHD (Plant Homeodomain) finger: Involved in chromatin binding
Linker histone-like domain: Facilitates nucleosome interactions
These domains work together to enable SHPRH's dual functionality in DNA repair and chromatin regulation. Unlike typical SNF2-family enzymes, SHPRH's ATPase activity does not translate into conventional nucleosome remodeling under standard assay conditions, suggesting specialized functions related to ubiquitination rather than chromatin restructuring .
SHPRH demonstrates distinctive binding preferences for chromatin components. Electrophoretic mobility shift assays (EMSAs) have revealed that:
SHPRH binds equally well to double-stranded DNA and nucleosome core particles
SHPRH shows a strong preference for nucleosomes presenting extranucleosomal DNA
Binding to nucleosomes with extranucleosomal DNA (147-bp core + 80-bp protruding DNA) occurs at significantly lower SHPRH concentrations compared to free DNA or core particles alone
This binding behavior resembles that of ISWI- and CHD-family chromatin remodeling factors, which also preferentially bind to nucleosomes with extranucleosomal DNA. The enhanced binding to these structures suggests that SHPRH may primarily function at nucleosomes during DNA repair processes or transcriptional regulation where partially unwrapped nucleosomes are present .
Circular RNA derived from the SHPRH gene (circ-SHPRH) has emerged as a potential biomarker and tumor suppressor distinct from the SHPRH protein itself. Key findings include:
Mechanistically, circ-SHPRH acts as a miRNA sponge, regulating downstream genes and signaling pathways that affect cancer cell proliferation, invasion, and apoptosis. This suggests separate but potentially complementary roles for SHPRH protein and circ-SHPRH in maintaining genomic stability and preventing tumorigenesis .
To accurately measure SHPRH's ATPase activity, researchers should implement the following methodological approach:
Reaction Components:
Purified recombinant SHPRH (50-100 nM)
ATP (0.1-1 mM)
Nucleosomes with extranucleosomal DNA (optimal substrate based on binding studies)
Magnesium ions (2-5 mM) as cofactor
Buffer system (typically Tris-HCl pH 7.5-8.0, 50-100 mM NaCl, 1 mM DTT)
Measurement Methods:
Colorimetric phosphate detection (e.g., malachite green assay)
Coupled enzyme assay using pyruvate kinase and lactate dehydrogenase
Radioactive ATP hydrolysis assay with [γ-³²P]ATP
Critical Controls:
DNA-only reactions
Nucleosome core particles without extranucleosomal DNA
No-ATP controls to establish baseline
Time course measurements to determine linear range
Research has demonstrated that nucleosomes strongly stimulate SHPRH's ATPase activity compared to free DNA, with nucleosomes containing extranucleosomal DNA providing maximal stimulation. Unlike conventional chromatin remodeling enzymes, this ATPase activity does not result in canonical nucleosome repositioning, suggesting a specialized function likely related to facilitating SHPRH's E3 ligase activity .
Assessment of SHPRH's E3 ubiquitin ligase activity requires careful experimental design:
Reaction Setup:
Purified recombinant SHPRH
Ubiquitin (wild-type or tagged versions)
E1 activating enzyme (typically UBA1)
E2 conjugating enzymes (research indicates SHPRH works with at least 7 different E2s)
ATP regeneration system
Target substrate (nucleosomes, PCNA, or other candidate proteins)
Recommended E2 Enzymes:
The following table outlines key E2 enzymes shown to function with SHPRH:
| E2 Family | Specific Enzymes | Ubiquitination Efficiency |
|---|---|---|
| UBE2D family | UBE2D1, UBE2D2, UBE2D3 | High |
| UBE2E family | UBE2E1, UBE2E2, UBE2E3 | High |
| UBE2W | UBE2W | Moderate |
| Others | Testing recommended | Variable |
Detection Methods:
Western blotting with anti-ubiquitin antibodies
Mass spectrometry for ubiquitination site and chain type identification
Fluorescently tagged ubiquitin for in-gel detection
Mass spectrometry analyses have revealed that SHPRH working with UBE2D1 can catalyze the formation of diverse polyubiquitin chains, including:
Branched polyubiquitin linkages
Linkages associated with DNA repair factor recruitment
Linkages involved in proteasomal degradation (K48-linked)
This diversity of chain types indicates that SHPRH may have multiple functions in the DNA damage response pathway .
To characterize SHPRH's interaction with nucleosomes, researchers should consider:
Nucleosome Preparation:
Reconstitute nucleosomes with defined DNA sequences:
147 bp DNA for core particles
227 bp DNA (147 bp + 80 bp extension) for nucleosomes with extranucleosomal DNA
Consider incorporating modified histones to assess their effect on binding
Binding Assays:
Electrophoretic Mobility Shift Assay (EMSA):
Titrate increasing SHPRH concentrations with fixed nucleosome concentration
Run on native polyacrylamide gels
Detect shifts through fluorescence or staining
Fluorescence Anisotropy:
Use fluorescently labeled nucleosomes
Monitor changes in anisotropy upon SHPRH binding
Calculate binding constants
Pulldown Assays:
Immobilize tagged SHPRH or nucleosomes
Assess binding partners and stability
Data Analysis:
Plot binding curves to determine dissociation constants (Kd)
Compare binding affinities between different nucleosome constructs
Analyze binding stoichiometry
Research has demonstrated that SHPRH binding to nucleosomes containing extranucleosomal DNA forms more stable complexes than with either free DNA or nucleosome core particles alone. Quantification of EMSA experiments showed that binding to nucleosomes with extranucleosomal DNA occurred at much lower SHPRH concentrations, indicating higher affinity for these substrates compared to free DNA or core particles .
Understanding the interplay between SHPRH's ATPase and E3 ligase activities represents a frontier in current research. Available evidence suggests a complex relationship that can be investigated through:
Domain-Function Analysis:
Generate SHPRH variants with mutations in:
ATPase domain (Walker A/B motifs)
RING finger domain
PHD finger domain
Assess each variant for ATPase activity, E3 ligase activity, and nucleosome binding
Determine interdependence between domains
Reaction Coupling Studies:
Compare sequential versus simultaneous ATPase and ubiquitination reactions
Analyze reaction products using mass spectrometry
Determine if ATP hydrolysis enhances ubiquitination specificity or efficiency
Structural Analysis:
Perform structural studies of SHPRH in different nucleotide-bound states
Identify conformational changes that might coordinate the two activities
Map interaction surfaces for E2 enzymes and nucleosome binding
Current research indicates that while nucleosomes stimulate SHPRH's ATPase activity, this doesn't translate into conventional nucleosome remodeling. Instead, ATP hydrolysis may facilitate conformational changes in SHPRH that enhance its ubiquitination activity. The finding that SHPRH can self-ubiquitinate within functional protein domains suggests a potential autoregulatory mechanism that could coordinate these activities .
SHPRH demonstrates versatility in its ability to function with multiple E2-conjugating enzymes and ubiquitinate various substrates. Understanding the determinants of this specificity requires:
E2 Enzyme Specificity:
Perform comprehensive screening with diverse E2 enzyme panels
Identify structural features that determine E2-SHPRH compatibility
Characterize ubiquitination patterns resulting from different E2-SHPRH pairs
Substrate Recognition:
Compare ubiquitination efficiency on different substrates:
Free histones vs. nucleosomes
Nucleosomes with different DNA lengths
Nucleosomes with various histone modifications
Identify substrate recognition motifs or surfaces
Determine how nucleosome binding affects target specificity
Mass Spectrometry Analysis:
Analyze ubiquitination sites and chain types formed with different E2-SHPRH combinations:
| E2 Enzyme | Preferred Chain Types | Target Specificity |
|---|---|---|
| UBE2D1 | Diverse including K48, K63, branched | Broad specificity on histones |
| UBE2E family | To be determined | Requires investigation |
| UBE2W | N-terminal ubiquitination | Requires investigation |
Research has shown that SHPRH can recruit E2 enzymes such as UBE2D1 to nucleosomes and form stable complexes. This recruitment ability may contribute to SHPRH's specificity in chromatin contexts. Mass spectrometry analyses have indicated that SHPRH with UBE2D1 can ubiquitinate nucleosomes with broad specificity and generate diverse polyubiquitin chains .
While SHPRH's role in polyubiquitinating PCNA during DNA damage response is established, emerging evidence suggests broader functions:
Chromatin-level Repair Mechanisms:
Investigate SHPRH recruitment to different types of DNA damage:
UV-induced damage
Double-strand breaks
Interstrand crosslinks
Characterize changes in chromatin accessibility at damage sites
Identify additional substrates at damage sites
Nucleosome Ubiquitination:
Map ubiquitination sites on histones catalyzed by SHPRH
Determine whether these modifications:
Recruit specific repair factors
Affect chromatin structure
Promote nucleosome removal or exchange
Non-ubiquitination Functions:
Assess potential roles in:
Chromatin remodeling during repair
Stabilizing repair intermediates
Regulating access to damaged DNA
Research has proposed that beyond PCNA ubiquitination, SHPRH may promote DNA repair or transcriptional regulation through chromatin ubiquitination. Mass spectrometry analyses have revealed that SHPRH can catalyze formation of polyubiquitin linkages associated with the recruitment of DNA repair factors, suggesting a broader role in coordinating repair processes at the chromatin level .
Mass spectrometry has become essential for characterizing SHPRH's ubiquitination targets and patterns. Researchers should implement the following analytical approach:
Sample Preparation:
Perform in vitro ubiquitination reactions with SHPRH, E2 enzymes, and nucleosomal substrates
Digest samples with trypsin (creates signature peptides with diglycine remnants on ubiquitinated lysines)
Consider enrichment for ubiquitinated peptides using anti-K-ε-GG antibodies
Data Analysis Workflow:
Ubiquitination Site Identification:
Search for +114.0429 Da mass shift on lysine residues
Apply appropriate false discovery rate thresholds
Validate with manual spectrum inspection
Chain Linkage Analysis:
Identify signature diglycine-modified peptides from ubiquitin itself
Quantify relative abundance of each linkage type
Determine branch points by identifying peptides with multiple modifications
Interpretation Framework:
| Ubiquitin Linkage | Cellular Function | SHPRH-Generated Patterns |
|---|---|---|
| K48-linked | Proteasomal degradation | Present in SHPRH reactions |
| K63-linked | DNA repair signaling | Present in SHPRH reactions |
| Branched chains | Enhanced recognition | Detected in SHPRH reactions |
| Other linkages | Various signaling roles | Requires further characterization |
Research has shown that SHPRH working with UBE2D1 can generate polyubiquitin chains with various linkages, including those associated with DNA repair factor recruitment and those involved in proteasomal degradation. This diversity suggests that SHPRH may have multiple functions in the cellular response to DNA damage .
Identifying SHPRH genomic binding sites requires integrative bioinformatic approaches:
Sequence-Based Analysis:
Identify consensus sequences from known binding sites
Apply motif discovery algorithms to ChIP-seq or similar data
Generate position weight matrices for genome-wide prediction
Structural Feature Recognition:
Characterize DNA structural properties of known binding regions:
DNA shape parameters
Intrinsic curvature
Flexibility and deformability
Apply machine learning to identify similar regions genome-wide
Chromatin Context Integration:
Correlate potential binding sites with:
Nucleosome positioning data
Histone modification patterns
DNA accessibility profiles
Prioritize sites with favorable chromatin contexts
Validation Experiments:
Perform ChIP-qPCR on predicted high-confidence sites
Test binding affinity with in vitro assays
Assess functional relevance through genetic perturbation
Research has shown that SHPRH preferentially binds nucleosomes with extranucleosomal DNA, suggesting that binding sites in vivo may be located at nucleosome boundaries or regions with partial nucleosome unwrapping. This preference should be incorporated into bioinformatic models for predicting SHPRH binding sites with higher accuracy .
Literature on SHPRH and circ-SHPRH shows some variation across studies and cancer types. To address these apparent contradictions:
Systematic Data Evaluation:
Categorize findings by:
Experimental system (cell lines, tissues, animal models)
Analytical methods
Cancer type and stage
Molecular context (mutations, pathway alterations)
Identify sources of variability:
Technical versus biological variation
Context-dependent effects
Temporal dynamics
Integrated Analysis Approach:
| Finding Type | Consistent Observations | Variable Observations | Reconciliation Approach |
|---|---|---|---|
| circ-SHPRH expression | Downregulated in most cancers | Degree of downregulation varies | Quantify context-specific effects |
| Prognostic value | Generally associated with better outcomes | Effect size varies by cancer type | Meta-analysis with subgroup analysis |
| Molecular mechanism | Acts as miRNA sponge | Different miRNA targets in different cancers | Map cancer-specific regulatory networks |
Cross-validation Strategy:
Test hypotheses across multiple model systems
Employ orthogonal experimental approaches
Perform tissue-specific analyses
SHPRH's roles in DNA repair and cancer suppression suggest potential therapeutic strategies:
Targeting Approaches:
Small Molecule Modulators:
ATP-competitive inhibitors targeting the ATPase domain
Allosteric modulators affecting E3 ligase activity
Protein-protein interaction disruptors targeting E2-SHPRH interfaces
Stabilization Strategies:
Compounds that prevent SHPRH degradation
Agents that enhance SHPRH expression or activity
Drugs targeting negative regulators of SHPRH
Cancer-Specific Considerations:
| Cancer Type | SHPRH/circ-SHPRH Status | Therapeutic Approach | Mechanistic Basis |
|---|---|---|---|
| Renal Cell Carcinoma | SHPRH stabilization beneficial | Develop axitinib-like compounds | Enhanced β-catenin degradation |
| Hepatocellular Carcinoma | circ-SHPRH downregulated | Restore circ-SHPRH expression | miRNA sponging activity |
| Multiple cancer types | circ-SHPRH as biomarker | Diagnostic and monitoring | Early detection and treatment selection |
Research has shown that axitinib, a tyrosine receptor kinase inhibitor used for renal cell carcinoma, appears to act partly by stabilizing SHPRH, which in turn increases ubiquitination and degradation of β-catenin, a central coactivator of oncogenic Wnt-responsive genes. This mechanism suggests that modulating SHPRH activity could be therapeutically beneficial in cancers with hyperactive Wnt signaling .
Despite promising potential, several challenges exist in targeting SHPRH therapeutically:
Biological Challenges:
Dual Enzymatic Functions:
Distinguishing between ATPase and E3 ligase activities
Selectively targeting one function while preserving the other
Understanding pathway-specific roles
Context-Dependent Activities:
Tissue-specific functions
Cancer-type variations
Genetic background effects
Technical Challenges:
Structural Complexity:
Large multi-domain protein difficult to express
Complex conformational dynamics
Multiple interaction surfaces
Selectivity Issues:
Similarity to other SNF2 family members
E3 ligase domain homology with other RING proteins
Potential off-target effects
Strategic Approaches:
Conduct comprehensive structural characterization:
Apply AI-driven conformational ensemble generation
Identify unique binding pockets
Develop highly selective modulators
Exploit cancer-specific vulnerabilities:
Synthetic lethal approaches
Tumor-specific expression patterns
Combination therapies
Advanced AI algorithms have been applied to predict alternative functional states of SHPRH, including large-scale conformational changes, to provide a robust foundation for structure-based drug design. These approaches may help overcome the challenges associated with targeting this complex protein .
Circ-SHPRH shows promise as a cancer biomarker based on consistent downregulation across multiple cancer types:
Biomarker Development Pathway:
Analytical Validation:
Standardize detection methods (qRT-PCR, RNA-seq)
Establish reference ranges in healthy tissues
Determine assay sensitivity and specificity
Clinical Validation:
Correlate with disease stage and progression
Assess prognostic value in prospective studies
Compare with existing biomarkers
Clinical Utility:
Develop minimally invasive testing (liquid biopsy)
Establish decision algorithms
Implement in therapeutic selection
Performance Characteristics: