Recombinant Xenopus laevis Cysteine and histidine-rich protein 1-A (cyhr1-a)

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your preferred format in your order notes for customized preparation.
Lead Time
Delivery times vary depending on the purchasing method and location. Please contact your local distributor for precise delivery estimates.
Note: Our proteins are shipped with standard blue ice packs. Dry ice shipping requires prior arrangement and incurs additional charges.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to collect the contents. Reconstitute the protein in sterile deionized water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our standard glycerol concentration is 50%, which can serve as a guideline.
Shelf Life
Shelf life depends on several factors including storage conditions, buffer composition, temperature, and the protein's inherent stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized forms have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquot to prevent repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing.
The tag type will be determined during the production process. If you require a specific tag, please inform us, and we will prioritize its development.
Synonyms
cyhr1-aCysteine and histidine-rich protein 1-A
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-365
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Xenopus laevis (African clawed frog)
Target Names
cyhr1-a
Target Protein Sequence
MSEEREAPGP LASSSAGLGA EVGQEEVPGG AGPARLLLLP SDSDGPPKKR LRSEAEPGSV RLEERLYSVL CCTVCLDLPK ASVYQCTNGH LMCAGCFIHL LADSRLKEEQ ATCPNCRCEI SKSLCCRNLA VEKAVSELPS DCGFCLKQFP RSLLERHKKE ECQDRVTQCK YKRIGCPWEG PYHELTVHES ECCHPTKTGN ELMEILDEMD QTHKKEMQLY NSIFSLLSFE KIGYTEVQFR PYRTDDFITR LYYETPRFTV LNQTWVLKAR VNDSERNPNL SCKRTLSFQL ILKSKINSPM ECSFLLLKGP YDDVKIHPVI YHFVFTNENN ETEYVPLPII DSVECNKLLA AKNINLRLFI FQIQK
Uniprot No.

Target Background

Database Links

UniGene: Xl.84627

Protein Families
CYHR1 family
Subcellular Location
Cytoplasm.

Q&A

What is Xenopus laevis CYHR1-A and what makes it a valuable research target?

CYHR1-A is the Xenopus laevis ortholog of the cysteine and histidine-rich protein family. Similar to its human counterpart, CYHR1-A likely contains domains rich in cysteine and histidine residues that may participate in metal ion coordination and protein-protein interactions. The protein holds significance for comparative studies due to Xenopus laevis being a well-established model organism with characterized developmental stages and a sequenced genome .

The protein's value stems from several factors:

  • Its cysteine-rich domains potentially form critical disulfide bonds that influence protein structure and function, similar to what has been observed in other Xenopus proteins like XCGL-1

  • Its potential role in cellular processes like cytoskeletal organization, as Xenopus models have contributed extensively to understanding cytoskeletal components

  • The opportunity it provides for evolutionary insights when compared to mammalian CYHR1 proteins

How does Xenopus laevis serve as an effective model system for CYHR1-A studies?

Xenopus laevis offers multiple advantages as a model system for studying CYHR1-A:

  • Rapid development and easy manipulation: Cultures can be obtained quickly (approximately one day after fertilization), compared to other model systems that require longer development periods

  • Ex utero fertilization: This allows for easy access to embryos at various developmental stages

  • Well-characterized genome: The sequenced Xenopus genome enables precise genetic manipulation and analysis

  • Cellular visualization advantages: Xenopus cells possess relatively large structures (like growth cones) compared to other vertebrates, facilitating detailed imaging of cytoskeletal and other cellular components

  • Maintenance ease: Xenopus cell cultures can be maintained without special conditions for extended periods, making them ideal for long-term imaging studies

What are the key differences between CYHR1-A and other cysteine-rich proteins in Xenopus laevis?

While specific comparative data for CYHR1-A is limited in the search results, we can draw insights from other cysteine-rich proteins in Xenopus:

  • Unlike XCGL-1 (Xenopus cortical granule lectin-1), which forms disulfide-linked oligomers via specific cysteine residues (C18 and C35) and functions in fertilization membrane development, CYHR1-A likely has distinct cysteine residue patterns that serve different structural purposes

  • CYHR1-A may not share the galactose-binding properties characteristic of XCGL-1, as their functional domains differ despite both containing cysteine-rich regions

  • Based on human CYHR1 research, CYHR1-A may be involved in cellular proliferation and migration pathways rather than the carbohydrate recognition functions seen in XCGL-1

What basic protocols are recommended for initial characterization of recombinant CYHR1-A?

Initial characterization should include:

  • Expression system selection: Based on experience with other Xenopus proteins, HEK293T mammalian cells or Trichoplusia ni insect cells are recommended expression systems for maintaining proper post-translational modifications

  • Protein purification strategy:

    • Affinity chromatography (likely utilizing His-tag or similar purification tag)

    • Size exclusion chromatography to separate oligomeric states

    • Ion exchange chromatography for final purification steps

  • Structural analysis:

    • SDS-PAGE under reducing and non-reducing conditions to assess disulfide bond formation

    • Western blotting to confirm protein identity

    • Electron microscopy for preliminary structural characterization, similar to the four-lobed structure observed for XCGL-1

  • Basic functional assays:

    • Binding assays to identify potential interaction partners

    • Assessment of calcium or other metal ion dependencies

    • Preliminary cellular localization studies using fluorescently tagged CYHR1-A

What expression systems provide optimal yield and proper folding for recombinant Xenopus CYHR1-A?

The choice of expression system significantly impacts recombinant CYHR1-A quality and functionality. Based on experience with other cysteine-rich Xenopus proteins:

Expression SystemAdvantagesDisadvantagesRecommended Applications
HEK293T Mammalian Cells- Proper post-translational modifications
- Mammalian chaperones for folding
- Allows for disulfide formation
- Lower yield
- Higher cost
- Longer cultivation time
- Structural studies
- Functional assays
- Interaction studies
Trichoplusia ni Insect Cells- Higher expression levels
- Eukaryotic processing
- Good compromise of yield/function
- Some glycosylation differences
- Moderate cost
- Mass spectrometry
- Antibody production
- Biochemical characterization
E. coli- Highest yield
- Lowest cost
- Rapid growth
- Requires refolding from inclusion bodies
- Lacks post-translational modifications
- Potential endotoxin contamination
- Initial screening
- Domain studies
- Mutational analysis

For CYHR1-A, HEK293T or Trichoplusia ni cells are likely preferred for functional studies, as demonstrated with XCGL-1 where both systems allowed proper formation of disulfide-linked oligomers . For studies focusing on specific domains, E. coli expression with subsequent refolding might be sufficient, though careful optimization of refolding conditions would be critical.

How can researchers effectively analyze CYHR1-A disulfide patterns and their functional significance?

Analyzing disulfide bonds in cysteine-rich proteins like CYHR1-A requires a multi-faceted approach:

  • Site-directed mutagenesis: Systematically replace individual cysteine residues with alanine (Cys→Ala mutations) to identify those critical for intermolecular and intramolecular disulfide formation. This approach successfully identified C18 and C35 as essential for XCGL-1 oligomerization .

  • Non-reducing vs. reducing SDS-PAGE: Compare migration patterns under these conditions to visualize disulfide-dependent oligomeric states .

  • Mass spectrometry analysis:

    • Perform tryptic digestion under non-reducing conditions

    • Use liquid chromatography-tandem mass spectrometry (LC-MS/MS) to identify disulfide-linked peptides

    • Consider stable isotope labeling to enhance detection

  • Biochemical validation:

    • Ellman's reagent to quantify free thiol groups

    • Differential alkylation with iodoacetamide/iodoacetic acid

    • Circular dichroism spectroscopy to assess structural changes upon reduction

  • Functional correlation:

    • Compare activity of wild-type and cysteine mutants to correlate specific disulfide bonds with function

    • Assess protein stability under reducing conditions

By applying these techniques, researchers can construct a comprehensive disulfide map for CYHR1-A and determine how these bonds contribute to protein structure and function, similar to the approach that revealed the role of specific cysteines in XCGL-1 oligomerization .

What advanced imaging techniques are most suitable for studying CYHR1-A localization and dynamics in Xenopus cells?

Xenopus laevis offers unique advantages for imaging studies due to its relatively large cellular structures and ease of culture maintenance . For CYHR1-A visualization:

  • Live-cell fluorescence microscopy:

    • CYHR1-A can be tagged with fluorescent proteins (GFP, mCherry) to monitor dynamics

    • Xenopus cultures are ideal for long periods of live imaging without special conditions

    • Time-lapse microscopy can track CYHR1-A movement relative to cytoskeletal elements

  • Super-resolution microscopy techniques:

    • Stimulated emission depletion (STED) microscopy

    • Stochastic optical reconstruction microscopy (STORM)

    • Photoactivated localization microscopy (PALM)

    • These techniques can resolve CYHR1-A distribution beyond the diffraction limit of conventional microscopy

  • Correlative light and electron microscopy (CLEM):

    • Combines fluorescence imaging with the high resolution of electron microscopy

    • Particularly useful for detailed structural analysis, similar to how electron microscopy revealed the four-lobed structure of XCGL-1

  • Förster resonance energy transfer (FRET):

    • Can detect CYHR1-A interactions with potential binding partners

    • Useful for studying dynamic changes in protein conformation

  • Fluorescence recovery after photobleaching (FRAP):

    • Measures CYHR1-A mobility and turnover in different cellular compartments

    • Particularly valuable in Xenopus growth cones, which are larger than in other vertebrates

These techniques leverage the advantages of Xenopus cells while providing detailed insights into CYHR1-A behavior under various conditions and stimuli.

How should researchers design binding assays to characterize CYHR1-A interactions with potential partners?

Based on methodologies used for other Xenopus proteins, a comprehensive binding analysis would include:

  • Bio-Layer Interferometry (BLI):

    • Immobilize biotinylated ligands on streptavidin-coated biosensor tips

    • Measure association (ka) by dipping into CYHR1-A solution

    • Measure dissociation (kd) by transferring to buffer solution

    • Calculate binding kinetics parameters and KD values

    • This approach was successfully used with XCGL-1 to determine binding affinities to various ligands

  • Surface Plasmon Resonance (SPR):

    • Similar to BLI but with different detection principles

    • Allows real-time, label-free detection of molecular interactions

    • Determine association/dissociation rates and equilibrium constants

  • Isothermal Titration Calorimetry (ITC):

    • Measures heat changes during binding to determine:

      • Binding stoichiometry

      • Binding affinity (KD)

      • Thermodynamic parameters (ΔH, ΔS, ΔG)

  • Pull-down assays and co-immunoprecipitation:

    • Use tagged CYHR1-A to identify binding partners from cell lysates

    • Confirm interactions through reverse pull-downs

    • Validate with western blotting

  • Yeast two-hybrid screening:

    • Particularly relevant as CYHR1 was first discovered through yeast two-hybrid with galectin-3

    • Can identify novel interaction partners from cDNA libraries

Results should be presented as comprehensive binding kinetics tables including ka, kd, and KD values for each interaction pair, similar to how XCGL-1 binding parameters were reported .

How can CYHR1-A research contribute to understanding cytoskeletal dynamics in Xenopus laevis?

Xenopus laevis has proven invaluable for studying cytoskeletal components including actin, microtubules, and neurofilaments . CYHR1-A research can extend this understanding by:

  • Investigating CYHR1-A and cytoskeletal protein interactions:

    • Co-localization studies with actin, microtubules, and intermediate filaments

    • FRET-based interaction analysis in live cells

    • Immunoprecipitation studies to identify cytoskeletal binding partners

  • Assessing effects of CYHR1-A manipulation on cytoskeletal organization:

    • Knockdown studies using siRNA (similar to CYHR1 knockdown in TE-8 cells)

    • Overexpression of wild-type and mutant CYHR1-A

    • Live imaging of cytoskeletal dynamics in Xenopus growth cones after CYHR1-A manipulation

  • Exploring CYHR1-A in growth cone dynamics:

    • Xenopus has relatively large growth cones ideal for cytoskeletal imaging

    • Time-lapse microscopy to correlate CYHR1-A localization with growth cone movement

    • Analysis of growth cone turning responses in CYHR1-A-depleted neurons

  • Investigating CYHR1-A in cellular migration:

    • Track migration patterns in normal and CYHR1-A-modified cells

    • Assess focal adhesion dynamics and turnover rates

    • Quantify migration velocity, directionality, and persistence

This research could potentially reveal whether CYHR1-A functions similarly to human CYHR1, which affects cell proliferation and invasion in cancer contexts , perhaps through cytoskeletal regulation mechanisms.

What experimental approaches can resolve contradictory findings about CYHR1-A function?

When faced with contradictory results regarding CYHR1-A function, researchers should:

  • Validate protein identity and integrity:

    • Confirm sequence through mass spectrometry

    • Verify proper folding through circular dichroism

    • Ensure disulfide bond formation through non-reducing SDS-PAGE

    • Validate activity through functional assays

  • Control experimental variables:

    • Standardize expression systems (HEK293T vs. Trichoplusia ni vs. E. coli)

    • Compare native vs. recombinant proteins

    • Control for tag effects (His-tag, GST-tag, etc.)

    • Ensure consistent buffer conditions, especially calcium concentrations which affect many protein functions

  • Adopt complementary methodologies:

    • Combine in vitro biochemical assays with in vivo studies

    • Use both gain-of-function and loss-of-function approaches

    • Apply both immunological detection and fluorescent protein tagging

    • Validate findings across different Xenopus developmental stages

  • Consider context-dependent functions:

    • Test CYHR1-A function in different cell types

    • Examine developmental stage-specific effects

    • Investigate potential binding partners that may modulate function

  • Create a systematic data integration approach:

    • Document experimental conditions thoroughly

    • Create comparison tables highlighting methodological differences

    • Perform meta-analysis of findings across studies

    • Conduct blind replication studies with standardized protocols

This multifaceted approach can help reconcile seemingly contradictory findings by identifying the specific conditions under which certain CYHR1-A functions are observed.

How does CYHR1-A expression correlate with developmental stages in Xenopus laevis?

Understanding CYHR1-A expression patterns throughout Xenopus development requires systematic analysis:

  • Temporal expression analysis:

    • RT-qPCR to quantify CYHR1-A mRNA levels across developmental stages

    • Western blotting to assess protein levels

    • Consider using standardized developmental staging criteria for Xenopus laevis

  • Spatial expression patterns:

    • In situ hybridization to localize CYHR1-A mRNA in embryos

    • Immunohistochemistry with validated antibodies

    • Fluorescent reporter constructs under CYHR1-A promoter control

  • Single-cell expression profiling:

    • Single-cell RNA sequencing at key developmental transitions

    • Analysis of cell type-specific expression patterns

    • Correlation with known developmental markers

  • Functional significance assessment:

    • Targeted knockdown at specific developmental stages

    • Phenotypic analysis following stage-specific manipulation

    • Correlation with developmental events (similar to how XCGL-1 is associated with fertilization membrane formation)

  • Comparative analysis:

    • Expression patterns of CYHR1-A vs. human CYHR1

    • Correlation with expression of potential interaction partners

    • Comparison with other cysteine-rich proteins in Xenopus

A comprehensive developmental expression profile could provide important clues about CYHR1-A function and help connect its molecular properties with specific developmental processes.

What roles might CYHR1-A play in cellular signaling pathways?

Based on what is known about human CYHR1 and its discovery through interaction with galectin-3 , CYHR1-A may participate in various signaling pathways:

  • Potential involvement in proliferation signaling:

    • Human CYHR1 knockdown suppresses proliferation in cancer cells

    • CYHR1-A may similarly influence Xenopus cell proliferation

    • Experimental approach: BrdU incorporation assays in normal and CYHR1-A-depleted cells

  • Migration and invasion pathways:

    • Human CYHR1 knockdown reduces invasion activity

    • CYHR1-A might regulate migration through cytoskeletal interactions

    • Experimental approach: Wound healing and transwell migration assays

  • Potential calcium-dependent signaling:

    • Many cysteine-rich proteins interact with calcium, as seen with XCGL-1

    • CYHR1-A may function as a calcium sensor or calcium-dependent regulator

    • Experimental approach: Calcium imaging in cells expressing fluorescently tagged CYHR1-A

  • Possible involvement in lectin-mediated signaling:

    • Given CYHR1's discovery through galectin-3 interaction

    • CYHR1-A might participate in carbohydrate-recognition pathways

    • Experimental approach: Glycan array screening similar to that performed for XCGL-1

  • Stress response signaling:

    • Cysteine-rich proteins often participate in redox signaling and stress responses

    • CYHR1-A might function as a redox sensor through its cysteine residues

    • Experimental approach: Assess CYHR1-A disulfide state changes under oxidative stress

Investigating these potential signaling roles would provide important insights into CYHR1-A function and its conservation across species.

What statistical methods are most appropriate for analyzing CYHR1-A expression and functional data?

Rigorous statistical analysis of CYHR1-A data requires:

  • For expression studies:

    • Two-way ANOVA to assess effects of developmental stage and tissue type

    • Post-hoc tests (Tukey's HSD or Bonferroni) for multiple comparisons

    • Pearson correlation for associating expression with quantitative variables

    • Principal component analysis to identify key factors driving expression variation

  • For binding kinetics data:

    • Non-linear regression for curve fitting (association/dissociation curves)

    • Scatchard plot analysis for binding affinity

    • Statistical comparison of KD values across experimental conditions

    • Similar to approaches used for analyzing XCGL-1 binding parameters

  • For functional assays:

    • Student's t-test or Mann-Whitney U test for comparing two conditions

    • ANOVA with appropriate post-hoc tests for multiple conditions

    • Cox proportional hazards model for survival analysis (if applicable)

    • Similar to statistical approaches used in human CYHR1 studies

  • For imaging data:

    • Pearson's or Mander's coefficients for co-localization analysis

    • Kymograph analysis for dynamic protein movements

    • Mixed-effects models for time-series data

  • For multi-omic integration:

    • Network analysis to identify functional relationships

    • Machine learning approaches for pattern recognition

    • Meta-analysis techniques for integrating heterogeneous datasets

How can computational modeling enhance understanding of CYHR1-A structure-function relationships?

Computational approaches offer powerful insights into CYHR1-A properties:

  • Homology modeling:

    • Generate structural models based on human CYHR1 or other cysteine-rich proteins

    • Validate models through Ramachandran plots and energy minimization

    • Similar to molecular modeling approaches used for XCGL-1

  • Molecular dynamics simulations:

    • Assess structural stability under different conditions

    • Analyze conformational changes upon binding

    • Investigate the role of specific cysteine residues in structural integrity

  • Binding site prediction:

    • Identify potential ligand binding pockets

    • Calculate electrostatic surface potentials

    • Dock potential binding partners to predict interaction modes

  • Network analysis of protein-protein interactions:

    • Predict functional partners based on conserved interaction sites

    • Generate interactome maps to place CYHR1-A in cellular context

    • Compare networks across species to identify conserved functional modules

  • Machine learning approaches:

    • Train models to predict functional effects of mutations

    • Classify CYHR1-A within protein families based on sequence features

    • Identify structural patterns associated with specific functions

These computational methods can generate testable hypotheses about CYHR1-A function and guide experimental design, particularly when integrated with experimental data from binding studies and structural analysis.

What are the emerging technologies that may advance CYHR1-A research?

Several cutting-edge technologies hold promise for CYHR1-A research:

  • CRISPR-Cas9 genome editing in Xenopus:

    • Generate CYHR1-A knockout or knock-in models

    • Create tagged versions of endogenous CYHR1-A

    • Introduce specific mutations to test structure-function hypotheses

  • Cryo-electron microscopy:

    • Determine high-resolution structures of CYHR1-A complexes

    • Visualize conformational states

    • Similar to the electron microscopy approaches that revealed XCGL-1's four-lobed structure

  • Proximity labeling techniques:

    • BioID or TurboID fusions to identify proximal proteins in living cells

    • APEX2 for electron microscopy-compatible proximity labeling

    • Spatial mapping of CYHR1-A interactome

  • Single-molecule techniques:

    • FRET to measure conformational changes

    • Optical tweezers to assess mechanical properties

    • Total internal reflection fluorescence (TIRF) microscopy for membrane interactions

  • Integrative structural biology:

    • Combine X-ray crystallography, NMR, and cryo-EM data

    • Integrate with cross-linking mass spectrometry

    • Build comprehensive structural models

  • Protein engineering approaches:

    • Directed evolution to enhance specific functions

    • Domain swapping to test functional hypotheses

    • Creation of biosensors based on CYHR1-A

These technologies could overcome current limitations in understanding CYHR1-A structure, dynamics, and interactions, potentially revealing novel functions and applications.

How should researchers design experiments to distinguish between direct and indirect effects of CYHR1-A manipulation?

Distinguishing direct from indirect effects requires careful experimental design:

  • Time-course analysis:

    • Monitor changes at multiple timepoints after CYHR1-A manipulation

    • Immediate effects (minutes to hours) are more likely direct

    • Delayed effects (days) often reflect indirect consequences

  • Dose-dependency studies:

    • Establish clear dose-response relationships

    • Direct effects typically show proportional responses to manipulation level

    • Similar to approaches used in human CYHR1 knockdown studies

  • Rescue experiments:

    • Reintroduce wild-type CYHR1-A after knockdown

    • Test domain-specific contributions through truncation mutants

    • Validate specificity through rescue with orthologs

  • Direct binding validation:

    • In vitro reconstitution with purified components

    • Demonstrate direct physical interactions through multiple techniques

    • Similar to the binding kinetics studies performed with XCGL-1

  • Rapid induction systems:

    • Employ optogenetic or chemically-inducible systems for temporal control

    • Observe immediate consequences of activation/inactivation

    • Distinguish between acute and adaptive responses

  • Single-cell correlation analysis:

    • Correlate CYHR1-A levels with phenotypic outcomes at single-cell resolution

    • Direct effects show stronger correlations with CYHR1-A levels

    • Account for cell-to-cell variability

These approaches, particularly when used in combination, can provide strong evidence for distinguishing direct functional roles of CYHR1-A from secondary consequences of its manipulation.

What comparative studies between Xenopus CYHR1-A and mammalian CYHR1 would yield the most valuable insights?

Comparative studies could reveal conserved and divergent aspects of CYHR1 function:

  • Evolutionary analysis:

    • Phylogenetic reconstruction of CYHR1 proteins across species

    • Identification of conserved domains and species-specific features

    • Analysis of selection pressures on different protein regions

  • Functional complementation experiments:

    • Express Xenopus CYHR1-A in human cells with CYHR1 knockdown

    • Test if CYHR1-A can rescue phenotypes observed in human CYHR1 studies

    • Identify domains required for cross-species functionality

  • Comparative protein-protein interaction networks:

    • Perform interactome studies in both systems

    • Identify conserved and species-specific interaction partners

    • Determine whether interaction with galectin-3 is conserved

  • Structural comparison:

    • Generate structural models for both proteins

    • Analyze conservation of disulfide bonding patterns

    • Compare calcium-binding properties, if present

  • Expression pattern comparison:

    • Document tissue distribution in both species

    • Correlate expression with developmental and physiological processes

    • Identify conserved regulatory elements in promoter regions

These comparative approaches could reveal fundamental aspects of CYHR1 function conserved throughout evolution while highlighting adaptations specific to amphibian biology.

How might CYHR1-A research inform therapeutic approaches for human diseases involving CYHR1?

Given human CYHR1's association with cancer progression , CYHR1-A research could contribute to therapeutic development:

  • Structure-based drug design:

    • Use Xenopus CYHR1-A as a model for inhibitor development

    • Test compounds in both Xenopus and human systems

    • Identify binding pockets conserved across species

  • Functional domain mapping:

    • Determine which domains of CYHR1 are essential for cancer-promoting functions

    • Develop domain-specific inhibitors

    • Test peptide mimetics based on CYHR1-A sequences

  • Biomarker development:

    • Extend findings of CYHR1 as a prognostic marker

    • Develop assays based on CYHR1 protein or antibody detection

    • Correlate expression patterns with disease progression

  • Pathway-targeted therapeutics:

    • Identify downstream effectors of CYHR1 conserved between species

    • Develop combination therapeutic approaches

    • Target synthetic lethal interactions discovered through CYHR1-A research

  • Model system for preclinical testing:

    • Use Xenopus CYHR1-A modified cells to screen potential therapeutics

    • Leverage the advantages of Xenopus for imaging and biochemical studies

    • Develop high-throughput screening platforms

Translational research connecting Xenopus CYHR1-A findings to human CYHR1 could accelerate therapeutic development while providing fundamental insights into protein function.

What interdisciplinary approaches could reveal new dimensions of CYHR1-A biology?

Integrating multiple disciplines could open new avenues for CYHR1-A research:

  • Systems biology approaches:

    • Multi-omic integration (transcriptomics, proteomics, metabolomics)

    • Network modeling of CYHR1-A in cellular pathways

    • Perturbation analysis across multiple conditions

  • Developmental biology and regeneration:

    • Examine CYHR1-A in Xenopus tissue regeneration processes

    • Investigate potential roles in cellular reprogramming

    • Study expression during critical developmental transitions

  • Immunology perspectives:

    • Explore potential roles in amphibian immune responses

    • Investigate parallels with other cysteine-rich immune lectins like intelectins

    • Examine expression during immune challenges

  • Evolutionary developmental biology:

    • Compare CYHR1 function across diverse species

    • Analyze how CYHR1 function has been conserved or repurposed

    • Connect molecular function to species-specific adaptations

  • Biophysical approaches:

    • Atomic force microscopy to measure mechanical properties

    • Nuclear magnetic resonance to study protein dynamics

    • Single-molecule fluorescence to track conformational changes

These interdisciplinary approaches could place CYHR1-A research in broader biological contexts, potentially revealing unexpected functions and applications beyond current understanding.

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.