Recombinant Sheep Secreted phosphoprotein 24 (SPP2)

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 order notes for customized fulfillment.
Lead Time
Delivery times vary depending on the purchasing method and location. Please contact your local distributor for precise delivery estimates. Note: All proteins are shipped with standard blue ice packs. Dry ice shipping requires advance notice 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. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting at -20°C/-80°C. Our standard glycerol concentration is 50% and can be used as a reference.
Shelf Life
Shelf life depends on various factors including storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized formulations have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt. Aliquoting is essential for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing. Tag type is determined during production. If you require a specific tag, please inform us, and we will prioritize its development.
Synonyms
SPP2; SPP24; Secreted phosphoprotein 24; Spp-24; Secreted phosphoprotein 2
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
24-203
Protein Length
Full Length of Mature Protein
Purity
>85% (SDS-PAGE)
Species
Ovis aries (Sheep)
Target Names
SPP2
Target Protein Sequence
FPVYDYD PASLKEALSA SVAKVNSQSL SPYLFRAFRS SIKRVNALDE DSLTMDLEFR IQETTCRRES EADPATCDFQ RGYHVPVAVC RSTVRMSAER VQDVWVRCHW SSSSGSSSSE EMFFGDILGS STSRNSHLLG LTPDRSRGEP LYERSREMRR NFPLGNRRYS NPWPRARVNP GFE
Uniprot No.

Target Background

Function

May play a role in bone turnover regulation.

Database Links

KEGG: oas:443510

UniGene: Oar.1099

Protein Families
SPP2 family
Subcellular Location
Secreted.

Q&A

What is Sheep Secreted phosphoprotein 24 (SPP2) and what are its key structural characteristics?

Sheep Secreted phosphoprotein 24 (SPP2), also known as SPP24, is a secreted phosphoprotein with significant roles in biological processes. The protein structure features an N-terminal hydrophobic region and three conserved phosphatase-family motifs. SPP2 is predicted to function as an integral membrane protein with potentially up to 9 membrane-spanning segments .

Immunofluorescence microscopy typically reveals a reticular staining pattern concentrated in perinuclear and cytosolic regions of cells, with colocalization often observed with endoplasmic reticulum markers. The protein's molecular structure contributes significantly to its biological function and detection capabilities in research contexts .

How does Sheep SPP2 differ from other secreted phosphoproteins such as SPP1?

While both SPP1 and SPP2 belong to the secreted phosphoprotein family, they have distinct genomic positions, structures, and biological functions. SPP1 (Secreted phosphoprotein 1) has been associated with somatic cell count in milk production studies and has different genomic positioning compared to SPP2 .

The functional distinction is critical - SPP2 functions in different molecular pathways than SPP1, with research suggesting unique roles in cellular signaling cascades. Understanding these differences is essential for designing targeted experiments and correctly interpreting results when studying either protein independently .

What are the most reliable methods for detecting and quantifying Recombinant Sheep SPP2 in experimental samples?

The gold standard for detecting and quantifying Recombinant Sheep SPP2 in experimental samples is the sandwich ELISA (Enzyme-Linked Immunosorbent Assay) technique. This method employs antibodies specific for SPP2 pre-coated onto a microplate. When samples containing SPP2 are added, the protein binds to the immobilized antibody .

A typical detection protocol follows these steps:

  • Addition of samples to antibody-coated wells

  • Binding of SPP2 to the immobilized antibody

  • Addition of biotin-conjugated secondary antibody specific for SPP2

  • Addition of Streptavidin-HRP (Horseradish Peroxidase) complex

  • Addition of substrate solution to develop color proportional to SPP2 concentration

  • Measurement of color intensity to determine SPP2 concentration

This method offers high sensitivity and excellent specificity with minimal cross-reactivity or interference from similar compounds .

How can researchers optimize immunoassays for detecting low concentrations of SPP2 in complex biological matrices?

Optimizing immunoassays for SPP2 detection in complex matrices requires several methodological refinements:

  • Sample preparation optimization: Pre-clearing samples through centrifugation or filtration can reduce matrix interference. Consider using specific buffer compositions that minimize non-specific binding while preserving SPP2 integrity.

  • Antibody selection: Use high-affinity antibodies that specifically recognize sheep SPP2 epitopes. Validate antibody specificity using positive and negative controls to ensure they don't cross-react with similar proteins.

  • Signal amplification strategies: Implement biotin-streptavidin systems as they provide enhanced signal amplification compared to direct detection methods. For extremely low concentrations, consider using chemiluminescent substrates rather than colorimetric ones .

  • Extended incubation periods: Longer incubation times at controlled temperatures can improve sensitivity when detecting minimal SPP2 concentrations.

  • Standard curve optimization: Prepare standards in the same matrix as your samples to account for matrix effects on the assay performance.

These optimization approaches should be validated through precision testing (CV < 10%) and recovery experiments (80-120% range) to ensure reliability .

What are the key considerations for designing experiments involving Recombinant Sheep SPP2 in various research contexts?

When designing experiments involving Recombinant Sheep SPP2, researchers should address several critical factors:

  • Protein stability: SPP2 stability should be evaluated under different storage and experimental conditions. Determine optimal buffer compositions, temperature ranges, and handling procedures to maintain protein activity.

  • Sample collection and processing: Similar to methodologies employed in related protein studies, researchers should standardize collection protocols. For example, when collecting biological samples, consider using K3EDTA vacuum tubes for samples intended for downstream molecular analyses .

  • Appropriate controls: Include both positive and negative controls in experimental designs. For recombinant protein work, consider using:

    • Wild-type protein controls

    • Denatured protein controls

    • Vehicle-only controls

    • Species-specific controls if cross-species comparisons are involved

  • Statistical power calculation: Determine appropriate sample sizes based on expected effect sizes. In comparable studies of secreted phosphoproteins, populations of approximately 380 samples have provided adequate statistical power for association analyses .

  • Reproducibility considerations: Document all experimental procedures in detail, including protein concentrations, incubation times, and washing steps to ensure other researchers can replicate your findings .

These design considerations help establish scientific rigor while ensuring meaningful and interpretable results.

How should researchers approach sample collection and preservation to maintain SPP2 integrity for accurate analysis?

Sample collection and preservation for SPP2 analysis requires careful attention to methodological details:

  • Collection timing: Standardize the timing of collection relative to physiological states or experimental manipulations. In animal studies, record parameters such as parity and time after relevant physiological events (e.g., lambing in sheep studies) .

  • Temperature control: Maintain samples at 4°C immediately after collection until analysis or longer-term storage. Avoid repeated freeze-thaw cycles which can degrade protein structure.

  • Preservation additives: Consider using protease inhibitors in sample collection tubes to prevent enzymatic degradation of SPP2.

  • Storage containers: Use sterile plastic containers (200 mL capacity has been validated in previous studies) for liquid biological samples to maintain sample integrity .

  • Long-term storage: For extended storage, maintain samples at -80°C rather than -20°C to better preserve protein structure and activity.

  • Documentation: Record comprehensive sample metadata including collection time, processing delays, and any deviations from standard protocols to help interpret potential anomalies in results .

Following these methodological guidelines helps ensure that the SPP2 protein remains structurally and functionally intact throughout the experimental process.

How can functional assays be designed to investigate the biological activity of Recombinant Sheep SPP2?

Designing functional assays for Recombinant Sheep SPP2 requires careful consideration of its putative biological roles:

  • Phosphatase activity assays: Since SPP2 contains phosphatase-family motifs, researchers can design assays to measure phosphatase activity using commercially available fluorogenic or chromogenic substrates. The assay should measure enzymatic activity under varying conditions (pH, temperature, cofactor requirements) .

  • Cell-based functional assays: Researchers can explore:

    • Overexpression systems using mammalian expression vectors

    • CRISPR-Cas9 knockout/knockdown approaches

    • Treatment of cultured cells with purified recombinant SPP2

    • Monitoring changes in cellular phenotypes (morphology, proliferation, migration)

  • Interaction studies: Investigate protein-protein interactions through:

    • Co-immunoprecipitation assays

    • Proximity ligation assays

    • Yeast two-hybrid screens

    • Surface plasmon resonance to quantify binding kinetics

  • Sphingolipid metabolism studies: Given the established connection between SGPP2 and sphingosine-1-phosphate (S1P) degradation, researchers should consider designing assays that monitor S1P levels in the presence of recombinant SPP2 under various experimental conditions .

These methodological approaches allow researchers to comprehensively characterize the functional properties of Recombinant Sheep SPP2 in diverse biological contexts.

What are the current challenges and limitations in studying the biological functions of Sheep SPP2?

Current research on Sheep SPP2 faces several methodological and conceptual challenges:

  • Limited cross-species validation: The degree to which findings from sheep SPP2 studies translate to other species remains unclear. Researchers must carefully design comparative studies that account for species-specific variations in protein structure and function.

  • Complex interactions with sphingolipid metabolism: The interaction between SPP2 and sphingolipid pathways creates methodological challenges in isolating specific effects attributable solely to SPP2 activity versus broader pathway perturbations .

  • Technical limitations in structural characterization: The membrane-associated nature of SPP2 presents challenges for structural biology techniques like X-ray crystallography and NMR spectroscopy, complicating efforts to fully elucidate structure-function relationships.

  • Variability in recombinant protein production: Expression systems may yield recombinant proteins with varying degrees of post-translational modifications compared to the native protein, potentially affecting functional studies.

  • Limited availability of sheep-specific research tools: Compared to human and mouse research, fewer validated antibodies, cell lines, and genetic models exist for sheep-specific research, creating methodological constraints.

Addressing these challenges requires innovative approaches and careful experimental design to advance understanding of Sheep SPP2 biology.

What methodologies are most appropriate for studying SPP2 gene expression in different sheep tissues?

Studying SPP2 gene expression in sheep tissues requires specialized methodological approaches:

  • Tissue sampling strategy: Implement a systematic sampling approach across multiple tissue types. Based on related studies, key tissues to examine include kidney, heart, brain, colon, lung, and small intestine, as these have shown variable expression levels of related phosphoproteins .

  • RNA extraction and quality control: Extract total RNA using established methods (TRIzol or column-based kits) followed by rigorous quality assessment (RNA integrity number > 7) before proceeding to expression analysis.

  • Quantitative expression analysis options:

    • RT-qPCR: Design primers specific to sheep SPP2 using available genomic information. Normalize expression using multiple reference genes validated for sheep tissues.

    • Northern blot analysis: For detection of full transcript size and splice variants. Previous studies have detected a 5.1-kb transcript in human tissues, providing a reference point for sheep studies .

    • RNA-seq: For global expression profiling and discovery of novel SPP2 splice variants.

  • In situ hybridization: To localize SPP2 mRNA expression within specific cell types in heterogeneous tissues.

  • Validation strategy: Confirm expression findings at the protein level using western blotting or immunohistochemistry with SPP2-specific antibodies.

These methods should be selected based on research objectives and available resources, with appropriate controls to ensure reliable and reproducible results.

How can researchers effectively analyze genetic polymorphisms of the SPP2 gene in sheep populations?

Analyzing genetic polymorphisms in the sheep SPP2 gene requires a structured methodological approach:

  • DNA sampling and extraction: Collect blood samples in K3EDTA vacuum tubes as demonstrated in previous sheep genetic studies. Extract DNA using standardized protocols that ensure high purity (A260/A280 ratio between 1.8-2.0) .

  • Polymorphism discovery approaches:

    • Targeted sequencing: PCR amplification and sequencing of exonic regions, splice junctions, promoter, and other regulatory regions.

    • Genome-wide approaches: Utilize whole-genome sequencing or SNP array technologies for broader discovery.

  • Genotyping methods: Select appropriate methods based on study scale:

    • PCR-RFLP (Restriction Fragment Length Polymorphism)

    • Real-time PCR with allele-specific probes

    • KASP (Kompetitive Allele Specific PCR) assays

    • Next-generation sequencing for high-throughput applications

  • Population genetics analysis: Calculate allele frequencies, genotype distributions, and test for Hardy-Weinberg equilibrium. Assess linkage disequilibrium patterns around the SPP2 locus.

  • Association analysis approach: Follow methods similar to those used in other sheep gene association studies:

    • Define phenotypes of interest clearly (e.g., production traits, disease resistance)

    • Account for population structure using principal component analysis

    • Apply appropriate statistical models (linear mixed models often preferred)

    • Implement rigorous correction for multiple testing

This systematic approach allows for comprehensive characterization of SPP2 genetic variation and its potential functional consequences in sheep populations.

How does Sheep SPP2 compare structurally and functionally with homologous proteins in other species?

Sheep SPP2 shows specific structural and functional relationships with homologous proteins across species:

SpeciesProtein Identity (%)Key Structural DifferencesFunctional Implications
Human~40% with SGPP1Different N-terminal region organizationPotentially altered membrane topology
BovineHigh (exact % varies by strain)Highly conserved phosphatase motifsSimilar enzymatic mechanisms
MurineModerateVariations in membrane-spanning segmentsPossible differences in subcellular localization
AvianLowSignificant divergence in hydrophobic regionsDistinct evolutionary adaptations

These comparative differences manifest in several functional aspects:

  • Substrate specificity: The conserved phosphatase motifs suggest similar catalytic mechanisms across species, but subtle structural differences may influence substrate binding affinities and kinetics.

  • Tissue expression patterns: Sheep SPP2 shows highest expression in kidney and heart, followed by brain, colon, lung, and small intestine, which parallels expression patterns observed in human tissues, suggesting evolutionary conservation of regulatory mechanisms .

  • Subcellular localization: The protein typically localizes to reticular patterns in perinuclear and cytosolic regions across species, with colocalization to the endoplasmic reticulum, indicating conserved trafficking mechanisms .

Understanding these comparative aspects provides valuable insights into the evolutionary conservation and divergence of SPP2 function across species.

What evolutionary insights can be gained from studying the SPP2 gene across different sheep breeds and related species?

Evolutionary analysis of the SPP2 gene offers valuable insights into adaptive processes and functional conservation:

  • Phylogenetic analysis methodology:

    • Sequence alignment of SPP2 genes from different sheep breeds and related species

    • Construction of phylogenetic trees using maximum likelihood or Bayesian methods

    • Calculation of evolutionary distances between sequences

  • Selection pressure analysis:

    • Calculate dN/dS ratios to identify regions under purifying or positive selection

    • Apply codon-based models to detect site-specific selection

    • Analyze conservation of functional domains across evolutionary history

  • Breed-specific variations:

    • Compare polymorphism patterns across traditional breeds (like Sarda sheep) and modern commercial breeds

    • Associate genetic variations with adaptations to specific environments or production traits

  • Domestication signatures:

    • Identify genetic changes that occurred during sheep domestication

    • Compare wild relatives to domestic breeds to pinpoint selection during the domestication process

  • Convergent evolution patterns:

    • Examine similar adaptations in SPP2 across unrelated species facing comparable environmental challenges

    • Identify parallel molecular adaptations that indicate functional importance

These evolutionary analyses provide context for understanding how SPP2 function has been shaped by natural and artificial selection, offering insights into its fundamental biological importance.

What role does SPP2 play in disease mechanisms, and how can this be effectively studied in sheep models?

SPP2's involvement in disease mechanisms can be investigated through systematic research approaches in sheep models:

  • Disease association methodology:

    • Conduct case-control studies comparing SPP2 expression and genetic variants between healthy and diseased sheep

    • Implement longitudinal studies tracking SPP2 levels before and during disease progression

    • Correlate SPP2 levels with established disease biomarkers

  • Mechanistic studies: Given SPP2's role in sphingolipid metabolism, particularly in S1P degradation, focus on inflammatory and immune-related conditions where S1P signaling is implicated :

    • Inflammatory diseases

    • Autoimmune conditions

    • Metabolic disorders

  • Gene manipulation approaches:

    • Develop transgenic sheep with SPP2 overexpression or knockdown

    • Utilize CRISPR-Cas9 for targeted gene editing in sheep cell lines or embryos

    • Apply RNA interference techniques in primary sheep cell cultures

  • Biomarker potential assessment:

    • Evaluate SPP2 as a diagnostic or prognostic biomarker using established ELISA methods

    • Determine sensitivity and specificity values for various disease conditions

    • Assess correlation with disease severity and treatment response

These methodological approaches enable comprehensive investigation of SPP2's role in pathological processes and its potential as a therapeutic target or biomarker.

How can recombinant Sheep SPP2 be utilized in developing novel therapeutic approaches?

Recombinant Sheep SPP2 offers several potential therapeutic applications that can be explored through structured research approaches:

  • Therapeutic protein development pipeline:

    • Optimize expression systems for producing functional recombinant Sheep SPP2

    • Develop purification protocols that maintain protein activity

    • Establish formulation parameters for stability and delivery

  • Mechanism-based therapeutic strategies:

    • Target SPP2's role in sphingolipid metabolism for conditions with dysregulated S1P signaling

    • Explore SPP2 supplementation in conditions with deficient endogenous levels

    • Develop SPP2 inhibitors for conditions with pathological overexpression

  • Delivery system optimization:

    • Evaluate various administration routes (intravenous, subcutaneous, tissue-specific)

    • Develop targeted delivery systems using nanoparticles or liposomes

    • Design sustained-release formulations for chronic conditions

  • Preclinical efficacy assessment:

    • Establish relevant animal models that recapitulate human pathologies

    • Define appropriate dosing regimens and treatment durations

    • Determine pharmacokinetic and pharmacodynamic profiles

  • Safety evaluation methodology:

    • Assess immunogenicity of recombinant Sheep SPP2

    • Evaluate toxicity profiles at therapeutic and supratherapeutic doses

    • Investigate potential off-target effects using proteomics and transcriptomics

These research directions provide a roadmap for translating basic SPP2 research into potential therapeutic applications while addressing critical methodological considerations.

What are the most effective mass spectrometry approaches for characterizing post-translational modifications of Sheep SPP2?

Characterizing post-translational modifications (PTMs) of Sheep SPP2 requires sophisticated mass spectrometry approaches:

  • Sample preparation optimization:

    • Employ enrichment strategies specific to the PTM of interest (e.g., TiO2 for phosphopeptides)

    • Use multiple proteases (trypsin, chymotrypsin, Glu-C) to maximize sequence coverage

    • Implement fractionation techniques (SCX, HILIC) to reduce sample complexity

  • MS instrumentation selection:

    • High-resolution instruments (Orbitrap, Q-TOF) are essential for accurate mass determination

    • Hybrid instruments allowing multiple fragmentation methods (CID, HCD, ETD) provide complementary information

    • Ion mobility MS can separate isomeric modified peptides

  • Data acquisition strategies:

    • Data-dependent acquisition for discovery of unknown PTMs

    • Parallel reaction monitoring for targeted quantification of known modifications

    • Data-independent acquisition for comprehensive PTM landscape analysis

  • Data analysis workflow:

    • Use multiple search engines (Mascot, SEQUEST, MS-GF+) with appropriate PTM settings

    • Apply PTM localization algorithms (PTM-score, Ascore) to determine exact modification sites

    • Implement false discovery rate control at both peptide and PTM-site levels

  • Quantitative analysis:

    • Label-free quantification for comparing PTM abundances across conditions

    • SILAC or TMT labeling for more accurate relative quantification

    • Absolute quantification using synthetic modified peptide standards

These mass spectrometry approaches enable comprehensive characterization of Sheep SPP2 PTMs, providing insights into their functional implications.

How can structural biology techniques be applied to understand the three-dimensional structure of Sheep SPP2?

Elucidating the three-dimensional structure of Sheep SPP2 requires an integrated structural biology approach:

  • X-ray crystallography strategy:

    • Protein expression optimization in bacterial, insect, or mammalian systems

    • Purification protocol development focusing on homogeneity and stability

    • Crystallization screening using sparse matrix and targeted approaches

    • Data collection at synchrotron facilities for high-resolution structures

    • Structure solution by molecular replacement or experimental phasing

  • NMR spectroscopy approach:

    • Isotopic labeling (15N, 13C, 2H) of recombinant Sheep SPP2

    • Optimization of sample conditions (buffer, pH, temperature) for spectral quality

    • Collection of multidimensional spectra for resonance assignment

    • Distance restraint gathering through NOE experiments

    • Structure calculation and refinement using specialized software

  • Cryo-electron microscopy method:

    • Sample preparation on specialized grids with vitrification

    • Data collection with direct electron detectors

    • Single particle analysis with 2D classification and 3D reconstruction

    • Model building and refinement against the EM density

  • Integrative structural biology:

    • Combine low-resolution techniques (SAXS, SANS) with high-resolution methods

    • Incorporate hydrogen-deuterium exchange mass spectrometry data

    • Use computational approaches (homology modeling, molecular dynamics) to complement experimental data

  • Functional interpretation:

    • Identify catalytic residues and substrate binding pockets

    • Map evolutionarily conserved regions onto the structure

    • Perform molecular docking to understand ligand interactions

    • Validate structural predictions through site-directed mutagenesis

This comprehensive structural biology approach provides valuable insights into the molecular mechanism of Sheep SPP2 function.

What statistical approaches are most appropriate for analyzing complex datasets involving SPP2 expression and function?

Analyzing complex SPP2 datasets requires sophisticated statistical approaches tailored to specific experimental designs:

  • Differential expression analysis:

    • For normally distributed data: ANOVA with appropriate post-hoc tests (Tukey's, Bonferroni)

    • For non-parametric data: Kruskal-Wallis with post-hoc Mann-Whitney U tests

    • For time-series data: repeated measures ANOVA or mixed-effects models

    • Apply appropriate transformations (log, Box-Cox) for skewed distributions

  • Association analysis for genetic studies:

    • Linear mixed models to account for population structure and relatedness

    • Family-based association tests for pedigree data

    • Correction for multiple testing using Bonferroni or false discovery rate methods

  • Multivariate analysis techniques:

    • Principal component analysis for dimension reduction

    • Partial least squares regression for relating SPP2 measurements to multiple dependent variables

    • Hierarchical clustering to identify patterns in expression data

  • Survival analysis approaches (for disease progression studies):

    • Kaplan-Meier curves with log-rank tests for comparing groups

    • Cox proportional hazards models to identify risk factors

    • Competing risk analysis when multiple outcome events are possible

  • Power analysis and sample size calculation:

    • A priori power calculations to determine required sample sizes

    • Post-hoc power analysis to interpret negative findings

    • Consider effect sizes observed in previous studies as a guide

These statistical approaches should be implemented with rigorous attention to assumptions and appropriate reporting of uncertainty measures.

How can researchers effectively address data inconsistencies and contradictions in SPP2 research?

Addressing data inconsistencies and contradictions in SPP2 research requires a systematic methodological approach:

  • Source evaluation framework:

    • Assess methodological differences between contradictory studies

    • Evaluate sample sizes and statistical power of conflicting results

    • Consider differences in experimental models, reagents, and protocols

    • Examine potential sources of bias in study design

  • Replication strategy:

    • Implement direct replication studies with increased statistical power

    • Use multiple methodological approaches to address the same question

    • Collaborate with independent laboratories for validation

    • Preregister study protocols to minimize reporting bias

  • Meta-analysis approach:

    • Conduct systematic reviews of available literature

    • Perform quantitative meta-analyses when sufficient studies exist

    • Use random-effects models to account for between-study heterogeneity

    • Conduct sensitivity analyses to identify influential studies or factors

  • Reconciliation techniques:

    • Develop integrative models that accommodate seemingly contradictory results

    • Identify contextual factors that may explain differences (species, tissue type, physiological state)

    • Consider non-linear relationships or threshold effects

    • Explore interaction effects that may resolve apparent contradictions

  • Transparent reporting practices:

    • Communicate limitations and uncertainties clearly

    • Report all results, including null findings

    • Share raw data and analysis code when possible

    • Discuss alternative interpretations of the data

These methodological approaches promote scientific rigor while advancing understanding despite apparent contradictions in the literature.

What emerging technologies and methodologies show promise for advancing SPP2 research?

Several cutting-edge technologies hold significant potential for advancing SPP2 research:

  • Single-cell analysis technologies:

    • Single-cell RNA sequencing to reveal cell-type-specific SPP2 expression patterns

    • Single-cell proteomics to detect SPP2 protein at the individual cell level

    • Spatial transcriptomics to map SPP2 expression within tissue architecture

    • Mass cytometry for simultaneous detection of SPP2 and dozens of other proteins

  • CRISPR-based technologies:

    • CRISPR activation/interference for precise modulation of SPP2 expression

    • Base editing for introducing specific mutations without double-strand breaks

    • Prime editing for precise genetic modifications with minimal off-target effects

    • CRISPR screens to identify genes interacting with SPP2

  • Advanced imaging techniques:

    • Super-resolution microscopy for visualizing SPP2 localization at nanoscale resolution

    • Label-free imaging methods for studying native SPP2 in living systems

    • Correlative light and electron microscopy for combining functional and structural information

    • Intravital microscopy for studying SPP2 dynamics in living animals

  • Systems biology approaches:

    • Multi-omics integration (genomics, transcriptomics, proteomics, metabolomics)

    • Network analysis to position SPP2 within broader signaling pathways

    • Computational modeling of SPP2-mediated processes

    • Machine learning for pattern discovery in complex SPP2-related datasets

  • Organoid and microphysiological systems:

    • Sheep-derived organoids for studying SPP2 in tissue-specific contexts

    • Organ-on-chip systems for investigating SPP2 function under controlled conditions

    • Bioprinting of 3D tissues with defined SPP2 expression patterns

These emerging technologies provide unprecedented opportunities to advance understanding of SPP2 biology at multiple scales of biological organization.

What are the most promising research questions that remain unanswered about Sheep SPP2?

Several critical research questions about Sheep SPP2 remain to be addressed through rigorous experimental approaches:

  • Functional genomics questions:

    • What are the key regulatory elements controlling SPP2 expression in different sheep tissues?

    • How do genetic variants in the SPP2 gene influence protein function and expression?

    • What is the evolutionary history of SPP2 across Ovis species and how has domestication shaped its genetics?

  • Structural biology inquiries:

    • What is the detailed three-dimensional structure of Sheep SPP2?

    • How does this structure relate to its catalytic function in sphingolipid metabolism?

    • What structural features determine substrate specificity and enzyme kinetics?

  • Cell biology investigations:

    • What is the precise subcellular localization of SPP2 in different sheep cell types?

    • How is SPP2 trafficking and secretion regulated in response to cellular stimuli?

    • What protein-protein interactions govern SPP2 function in different cellular compartments?

  • Physiological function questions:

    • What are the physiological consequences of SPP2 dysregulation in sheep?

    • How does SPP2 contribute to normal development and tissue homeostasis?

    • What compensatory mechanisms exist when SPP2 function is compromised?

  • Translational research priorities:

    • Can SPP2 serve as a biomarker for specific sheep diseases or conditions?

    • What is the therapeutic potential of recombinant SPP2 or SPP2 modulators?

    • How can insights from sheep SPP2 research inform human biomedical applications?

Addressing these questions requires innovative experimental designs, cutting-edge methodologies, and collaborative research efforts across disciplines.

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.