Recombinant Human Ankyrin Repeat Domain-Containing Protein 37 (ANKRD37) is a protein that contains ankyrin repeat domains, which are known for mediating protein-protein interactions. These interactions are crucial in various cellular processes, including cell signaling, transcription regulation, and cell cycle control . ANKRD37 is a protein-coding gene in humans and has been associated with several biological functions and diseases .
ANKRD37 contains four ankyrin repeats, which are structural motifs that facilitate protein-protein interactions . Ankyrin repeats are composed of two antiparallel α-helices followed by a β-hairpin or a long loop, forming an L-shaped domain that resembles a cupped hand . This structure allows ANKRD37 to interact with other proteins, potentially influencing transcriptional regulation and signaling pathways.
ANKRD37 has been identified as a novel target gene of Hypoxia-Inducible Factor-1 (HIF-1), suggesting its involvement in the cellular response to hypoxia . HIF-1 is a transcription factor that regulates gene expression under low oxygen conditions, and ANKRD37's induction in hypoxia indicates its potential role in adapting to such environments.
ANKRD37 has been linked to diseases such as myxosarcoma, highlighting its potential involvement in cancer biology . Additionally, research suggests a possible causal association between ANKRD37 and human hippocampal volume, indicating its role in neurological functions .
ANKRD37 expression can be influenced by various environmental and chemical factors. For example, certain chemicals like cobalt dichloride and atrazine increase ANKRD37 mRNA expression, while others such as benzo[a]pyrene and 2,3,7,8-tetrachlorodibenzodioxine decrease its expression . This suggests that ANKRD37's expression is sensitive to environmental stressors and toxic substances.
| Chemical/Substance | Effect on ANKRD37 Expression |
|---|---|
| Cobalt Dichloride | Increases |
| Atrazine | Increases |
| Benzo[a]pyrene | Decreases |
| 2,3,7,8-Tetrachlorodibenzodioxine | Decreases |
| Arsenic | Increases |
| 2-Palmitoylglycerol | Increases |
Note: We will prioritize shipping the format currently in stock. If you require a specific format, please specify this during order placement.
Note: All proteins are shipped with standard blue ice packs. Dry ice shipping requires prior arrangement and incurs additional charges.
The tag type will be determined during production. If you require a specific tag, please inform us, and we will prioritize its development.
ANKRD37 contains multiple ankyrin repeat motifs that fold into characteristic L-shaped domains. Each repeat consists of two antiparallel α-helices followed by a β-hairpin or extended loop. These repeats stack together to form a slightly curved structure resembling a cupped hand, where the β-hairpins represent the "fingers" and the α-helices form the "palm" . This architectural arrangement creates a specific protein-binding interface that mediates ANKRD37's interactions with partner proteins. Unlike globular proteins, ankyrin repeat domains are dominated by local, short-range interactions, which represents a distinct paradigm for protein stability and folding mechanisms .
While the search results don't specifically mention the number of repeats in ANKRD37, analysis of ankyrin repeat proteins shows that they typically contain between 1-33 repeats, with most proteins having six or fewer repeats . The detection of terminal repeats can be challenging due to sequence divergence from the consensus, particularly as terminal repeats often contain polar residues that facilitate interactions with solvent, replacing the well-conserved hydrophobic residues found in internal repeats .
Based on the known functions of ankyrin repeat proteins, ANKRD37 likely participates in cellular processes such as transcriptional regulation, signal transduction, or protein complex assembly. Ankyrin repeat proteins generally function as scaffolds for protein-protein interactions without enzymatic activity . Other ankyrin repeat proteins participate in cell-cell signaling, cytoskeleton integrity, transcription and cell-cycle regulation, inflammatory response, development, and various transport phenomena .
For analyzing ANKRD37 expression patterns, researchers should consider complementary approaches including:
RNA-based methods: qRT-PCR for quantitative mRNA expression analysis, RNA-seq for comprehensive transcriptomic profiling, and in situ hybridization for spatial localization in tissue sections.
Protein-based methods: Western blotting with ANKRD37-specific antibodies for protein quantification, immunohistochemistry/immunofluorescence for visualization in tissues, and mass spectrometry for proteomic analysis.
Reporter systems: Creation of ANKRD37 promoter-reporter constructs to monitor transcriptional regulation in different cell types or in response to various stimuli.
The combination of these techniques provides a comprehensive view of ANKRD37 expression patterns, essential for understanding its physiological roles.
To investigate ANKRD37 regulation, researchers could explore:
Transcription factor binding: Chromatin immunoprecipitation (ChIP) to identify transcription factors binding to the ANKRD37 promoter.
Epigenetic regulation: Bisulfite sequencing to analyze DNA methylation patterns and ChIP for histone modifications associated with the ANKRD37 gene.
Experimental manipulation strategies:
Gene silencing using siRNA or CRISPR/Cas9 systems
Overexpression using recombinant expression vectors
Treatment with chemical modulators of signaling pathways potentially regulating ANKRD37
Integrating these approaches provides a comprehensive understanding of ANKRD37 regulation mechanisms.
The choice of expression system depends on research objectives:
Bacterial expression (E. coli):
Advantages: High yield, simplicity, cost-effectiveness
Limitations: Potential for improper folding of ankyrin repeats due to lack of eukaryotic chaperones
Optimization: Use specialized strains with enhanced disulfide bond formation capabilities; expression at lower temperatures (16-18°C); fusion with solubility-enhancing tags (MBP, SUMO)
Insect cell systems:
Advantages: Eukaryotic folding machinery, post-translational modifications
Optimization: Baculovirus expression vector systems with optimized signal sequences and purification tags
Mammalian expression systems:
Advantages: Native folding environment, appropriate post-translational modifications
Systems: HEK293 or CHO cells for transient or stable expression
The selection should be guided by whether native conformation and post-translational modifications are critical for the planned experiments.
A multi-step purification strategy is recommended:
Initial capture:
Immobilized metal affinity chromatography (IMAC) for His-tagged constructs
Glutathione affinity chromatography for GST-fusion proteins
Intermediate purification:
Ion exchange chromatography based on ANKRD37's predicted isoelectric point
Heparin affinity chromatography if DNA/RNA binding properties are suspected
Polishing step:
Size exclusion chromatography to separate monomeric protein from aggregates and remove remaining impurities
Stability optimization:
Buffer screening using differential scanning fluorimetry
Addition of stabilizing agents (glycerol, specific salts, reducing agents)
The purification protocol should be validated using SDS-PAGE, Western blotting, and mass spectrometry to confirm identity and purity.
Multiple complementary techniques should be employed:
Circular dichroism (CD) spectroscopy: To assess secondary structure content and confirm the expected α-helical signature of ankyrin repeats
Thermal denaturation studies: To analyze the cooperative unfolding behavior characteristic of properly folded ankyrin domains. Previous studies on ankyrin repeat domains have shown they typically exhibit two-state folding transitions despite their modular structure
Limited proteolysis: Well-folded ankyrin repeat domains show characteristic resistance patterns to proteolytic digestion
Analytical ultracentrifugation: To confirm homogeneity and expected hydrodynamic properties
Functional assays: Verification of expected protein-protein interactions as the ultimate test of proper folding
Researchers should consider a multi-layered approach:
Screening methods:
Yeast two-hybrid screening
Mammalian two-hybrid systems
Affinity purification coupled with mass spectrometry (AP-MS)
Proximity-based labeling techniques (BioID, APEX)
Validation methods:
Co-immunoprecipitation in relevant cell types
GST pull-down assays with recombinant proteins
Surface plasmon resonance (SPR) for quantitative binding kinetics
Fluorescence resonance energy transfer (FRET) for interactions in living cells
The search results indicate numerous ankyrin repeat proteins have binding partners identified through these techniques, as shown in Table 1 from the literature .
This question addresses a fundamental property of ankyrin repeat domains:
Surface area effects: The number of ankyrin repeats directly impacts the available binding surface area. More repeats typically provide larger interaction interfaces, potentially increasing binding affinity and enabling multivalent interactions
Conformational considerations: The natural curvature of stacked ankyrin repeats creates a specific three-dimensional binding surface. This curvature becomes more pronounced with increasing repeat numbers, potentially affecting binding partner specificity
Experimental approaches to investigate this question:
Construct a series of ANKRD37 truncation mutants with varying numbers of repeats
Measure binding affinities (KD values) using techniques like isothermal titration calorimetry (ITC) or SPR
Analyze complex stability through thermal denaturation studies
Perform molecular dynamics simulations to model conformational effects
Studies on other ankyrin repeat proteins have demonstrated that deletion of C-terminal repeats can significantly impact stability, as shown in the Notch ankyrin domain where full seven-repeat constructs (Nank1-7*) had substantially higher stability than versions with fewer repeats .
Crystallization of ankyrin repeat proteins presents unique challenges and opportunities:
Construct optimization:
Systematic truncation series to identify stable, crystallizable fragments
Surface entropy reduction through mutation of high entropy residues (Lys, Glu) to alanine
Consideration of co-crystallization with binding partners to stabilize flexible regions
Crystallization conditions:
Initial broad screening followed by focused optimization
Attention to pH ranges 6.0-8.0, where ankyrin repeat proteins typically maintain stability
Inclusion of additives that promote crystal contacts without destabilizing the protein
Alternative approaches:
Fusion with crystallization chaperones (e.g., T4 lysozyme)
Synthetic ankyrin repeat proteins (DARPins) as crystallization aids
Successful crystallization of 13 natural and 3 designed ankyrin repeat proteins has been reported , providing precedent for structural studies of ANKRD37.
NMR approaches for ANKRD37 should consider:
Sample preparation considerations:
Expression of isotopically labeled protein (15N, 13C, 2H as needed)
Optimization of buffer conditions for NMR (avoidance of paramagnetic ions, pH control)
Potential segmental labeling for larger constructs
Experimental strategies:
Backbone assignment using TROSY-based experiments if molecular weight exceeds 20 kDa
Residual dipolar coupling measurements to define relative orientation of ankyrin repeats
Relaxation measurements to characterize dynamics of individual repeats
Data analysis approaches:
Automated assignment software combined with manual verification
Integration with previously solved ankyrin repeat structures as templates
Modeling of repeat-repeat interfaces
To investigate ANKRD37's role in cellular pathways:
Pathway analysis approaches:
Proteomics analysis following ANKRD37 knockdown/overexpression
Phosphoproteomics to identify signaling changes
Transcriptomics to identify genes regulated directly or indirectly by ANKRD37
Functional screening methods:
CRISPR/Cas9 screening to identify synthetic lethal interactions
High-content imaging following ANKRD37 perturbation
Protein-fragment complementation assays to map physical interaction networks
Integration with known ankyrin repeat protein functions:
Assessment of roles in established ankyrin repeat protein pathways such as:
Cell-cycle regulation (similar to INK4 proteins)
Transcriptional regulation (similar to IκB proteins)
Signal transduction (similar to Notch)
Cytoskeletal organization
The search results indicate ankyrin repeat proteins function in diverse cellular processes including cell-fate decisions, inflammatory responses, and cytoskeletal organization .
Detailed mapping of ANKRD37 binding interfaces requires:
Mutagenesis approaches:
Alanine scanning mutagenesis of surface residues
Charge reversal mutations at potential interaction sites
Creation of chimeric proteins by swapping repeats with other ankyrin repeat proteins
Structural analysis of complexes:
X-ray crystallography of ANKRD37 with binding partners
Cryo-EM for larger complexes
Crosslinking mass spectrometry to identify residues in proximity at binding interfaces
Computational methods:
Molecular docking guided by experimental constraints
Molecular dynamics simulations of protein-protein interactions
Previous co-crystal structures of ankyrin repeat proteins have shown that binding typically occurs at the concave inner surface comprising the β-hairpin/loop regions and inner short helices .
To investigate ANKRD37 in disease contexts:
Clinical correlation studies:
Analysis of ANKRD37 expression in patient samples using immunohistochemistry and qRT-PCR
Correlation of expression levels with disease progression and prognosis
Genotyping studies to identify disease-associated variants
Functional disease modeling:
Gene editing in cell lines using CRISPR/Cas9 to introduce disease-associated mutations
Patient-derived iPSCs differentiated into relevant cell types
Animal models with modified ANKRD37 expression or function
Mechanistic investigations:
Interactome analysis in disease vs. normal conditions
Signaling pathway perturbation studies
Drug screening to identify modulators of ANKRD37 function
Many ankyrin repeat proteins have been implicated in human diseases, with the INK4 family of tumor suppressors (p15, p16, p18, p19) being prominent examples .
For comprehensive characterization of disease-associated mutations:
Structural impact assessment:
In silico modeling of mutation effects on protein stability and conformation
Thermal denaturation studies comparing wild-type and mutant proteins
Limited proteolysis to detect conformational changes
Functional consequences evaluation:
Quantitative binding assays to assess effects on protein-protein interactions
Cellular localization studies using fluorescently tagged constructs
Functional complementation assays in knockout cell lines
Integrated approaches:
Creation of isogenic cell lines differing only in the mutation of interest
Multi-omics profiling to comprehensively characterize downstream effects
Phenotypic screening to identify cellular processes affected by mutations
Innovative protein engineering strategies include:
Designer ankyrin repeat proteins:
Creation of consensus-designed ankyrin repeat domains as scaffolds
Development of ANKRD37-based protein binders through directed evolution
Construction of chimeric proteins with repeats from different ankyrin proteins
Optogenetic and chemogenetic control:
Integration of light-sensitive domains to enable spatiotemporal control of ANKRD37 interactions
Creation of rapamycin-inducible dimerization systems to control ANKRD37 localization and function
Biosensor development:
Creation of FRET-based biosensors using ANKRD37 to monitor protein interactions in real-time
Development of split-protein complementation reporters based on ANKRD37 binding properties
Recent consensus-based protein design strategies have been successfully applied to ankyrin repeat proteins, leveraging the large number of available sequences .
Advanced computational methods include:
Sequence-based predictions:
Machine learning algorithms trained on known ankyrin repeat protein functions
Evolutionary analysis to identify conserved functional residues
Co-evolution analysis to predict protein-protein interactions
Structure-based approaches:
Molecular dynamics simulations to identify conformational states
Virtual screening for potential binding partners or small molecule modulators
Fragment-based computational design of interaction surfaces
Network-based methods:
Integration of protein-protein interaction networks, gene expression data, and phenotypic information
Graph theory approaches to predict functional associations
System-level modeling of ANKRD37 in cellular pathways
Evolutionary analysis of ANKRD37 can provide functional insights through:
Phylogenetic analysis methods:
Construction of phylogenetic trees to map ANKRD37 evolution
Identification of orthologs across diverse species
Analysis of repeat number variation across evolutionary lineages
Sequence conservation patterns:
Calculation of sequence conservation scores at binding interfaces versus non-interface regions
Identification of species-specific insertions or deletions
Detection of signatures of positive or negative selection
Functional divergence assessment:
Comparative expression pattern analysis across species
Cross-species complementation experiments
Comparison of binding partner networks in different organisms
The analysis of ankyrin repeat domains across species has revealed remarkable conservation of structure despite sequence variation, highlighting the evolutionary importance of this protein interaction module .
To assess functional conservation across species:
Heterologous expression studies:
Expression of ANKRD37 orthologs from different species in human cell lines
Assessment of subcellular localization patterns
Evaluation of protein-protein interaction profiles
Domain swapping experiments:
Creation of chimeric proteins with domains from ANKRD37 orthologs
Functional complementation testing in knockout systems
Binding affinity measurements with predicted conserved partners
Model organism studies:
CRISPR/Cas9 modification to humanize ANKRD37 in model organisms
Phenotypic comparison of mutants across species
Cross-species rescue experiments
| Species | Number of Ankyrin Repeats | Sequence Identity (%) | Key Conserved Interaction Motifs |
|---|---|---|---|
| Human | Variable (typically 4-7) | 100 (reference) | β-hairpin/loop regions, inner helices |
| Mouse | Similar to human | ~90-95 (estimated) | Conserved inner surface residues |
| Zebrafish | May vary | ~70-80 (estimated) | Core structural elements |
| Drosophila | Often fewer | ~40-60 (estimated) | Basic repeat architecture |
| C. elegans | Often fewer | ~30-50 (estimated) | Selected binding residues |
Note: This table presents typical patterns observed in ankyrin repeat proteins; specific values for ANKRD37 would require detailed sequence analysis.