Note: While we prioritize shipping the format currently in stock, please specify your preferred format in order notes for custom preparation.
Note: All proteins are shipped with standard blue ice packs unless otherwise requested. Dry ice shipping requires prior arrangement and incurs additional charges.
The tag type is determined during production. Specify your desired tag type in advance for preferential development.
Samd3 (Sterile alpha Motif Domain Containing 3) is characterized by the presence of a sterile alpha motif (SAM) domain, which is a protein interaction module of approximately 70 amino acids. The full-length mouse Samd3 protein consists of 520 amino acids with a molecular weight of approximately 61.2 kDa . The SAM domain is evolutionarily conserved and can mediate various protein-protein interactions, potentially facilitating the formation of homo- and hetero-oligomers with other SAM domain-containing proteins.
To properly investigate the structural properties of Samd3:
Begin with bioinformatic analysis using tools like SMART or Pfam to identify all functional domains
Validate domain predictions through recombinant protein expression studies with truncated constructs
Utilize circular dichroism spectroscopy to assess secondary structure characteristics
Consider X-ray crystallography or NMR spectroscopy for high-resolution structural determination
Samd3 belongs to the larger family of SAM domain-containing proteins, which includes other members like SAMD4B and SAMD5. While these proteins share the conserved SAM domain, they exhibit distinct expression patterns and functions. For instance, SAMD5 is specifically expressed in peribiliary glands (PBGs) of large bile ducts in mice and has been associated with cell cycle regulation in cholangiocarcinoma cell lines .
Comparative analysis methodology:
Perform sequence alignment of SAM domains across family members using multiple sequence alignment tools
Construct phylogenetic trees to visualize evolutionary relationships
Compare tissue expression profiles using public databases like GEO or ArrayExpress
Analyze protein-protein interaction networks to identify unique vs. shared interaction partners
Research has demonstrated that Samd3 exhibits a specific expression pattern within the immune system. Samd3 expression has been primarily observed in natural killer (NK) cells and CD8+ T cells, which are known for their specific functions against intracellular pathogens like viruses . Notably, after acute viral infections, Samd3 expression becomes enriched within memory precursor cells, and the frequency of Samd3-expressing cells increases during the memory development phase .
To accurately characterize Samd3 expression:
Utilize flow cytometry with fluorescent reporter mice (as described in the multifunctional mouse model)
Perform single-cell RNA sequencing of immune cell populations to capture heterogeneity
Validate with quantitative RT-PCR on sorted cell populations
Use immunohistochemistry to visualize expression in tissue contexts
During viral infection, Samd3 expression undergoes dynamic changes that correlate with specific T cell differentiation states. After acute viral infection, Samd3 expression becomes preferentially enriched in memory precursor cells, and the frequency of Samd3-expressing cells increases throughout the memory development phase .
In chronic viral infection scenarios, Samd3 expression is predominantly detected within precursors of exhausted CD8+ T cells, which are critical for viral control . This pattern suggests a potential role for Samd3 in the development or maintenance of specific T cell subsets during different types of immune responses.
Methodology for tracking expression changes:
Implement time-course studies using reporter mice infected with model viruses (e.g., LCMV)
Correlate Samd3 expression with established markers of T cell differentiation states
Perform RNA-seq analysis at different infection timepoints
Consider intravital imaging to track Samd3-expressing cells in vivo
Recombinant mouse Samd3 can be efficiently expressed using several expression systems, with mammalian expression systems like HEK-293 cells generally providing proper folding and post-translational modifications . For optimal expression and purification:
Expression System Selection:
HEK-293 cells yield well-folded protein with appropriate post-translational modifications
Cell-free protein synthesis (CFPS) systems can provide rapid production with >70-80% purity
Choose the system based on downstream applications and required protein quality
Tagging Strategy:
His-tag (6x histidine) enables purification via nickel affinity chromatography
Strep-tag facilitates gentle elution conditions and high specificity
For dual purification strategies, consider tandem tags separated by a protease cleavage site
Purification Protocol:
Begin with affinity chromatography based on the chosen tag
Follow with size exclusion chromatography to remove aggregates and achieve >90% purity
Validate purity using techniques such as Bis-Tris PAGE or SDS-PAGE
Confirm identity via Western blot with anti-tag antibodies or mass spectrometry
Storage Conditions:
Based on published approaches, researchers can develop a multifunctional mouse model to study Samd3 through these methodological steps:
Design Strategy:
Targeting Construct Components:
Homology arms flanking the Samd3 locus
loxP sites surrounding critical exons
Reporter gene under control of the endogenous Samd3 promoter
Selection markers for embryonic stem cell screening
Validation Approach:
Genotyping PCR to confirm correct integration
RT-PCR and Western blot to verify conditional knockout efficiency
Flow cytometry to validate reporter expression correlates with endogenous Samd3
Functional assays to confirm phenotypic changes after Cre-mediated recombination
Experimental Controls:
Include littermate controls without Cre recombinase
Use mice with Cre but without floxed Samd3 to control for Cre toxicity
Implement reporter-only controls to distinguish effects of gene deletion from cell depletion
While Samd3 expression is enriched in memory precursor cells and increases during memory development , its precise functional role remains incompletely understood. Current evidence suggests complex involvement that requires sophisticated experimental approaches:
Functional Assessment Methodology:
Use conditional knockout models with T cell-specific Cre drivers (CD8-Cre)
Implement adoptive transfer experiments with Samd3-deficient vs. wild-type T cells
Analyze cellular transcriptomes to identify differentially regulated pathways
Assess memory recall responses to secondary challenges
Research Considerations:
Examine both quantity (cell numbers) and quality (functional capacity) of memory T cells
Investigate both central memory and effector memory subsets
Evaluate memory T cell maintenance in bone marrow niches
Consider compensatory mechanisms from related SAM domain proteins
Contradictory Findings:
Current research presents an interesting contradiction: while Samd3 expression is enriched in memory precursor cells, Samd3-deficient CD8+ T cells were not functionally compromised in the context of acute infection with Vaccinia virus or chronic viral infections . This suggests several possibilities:
Redundancy with other SAM domain-containing proteins
Context-dependent functions requiring specific inflammatory environments
Subtle effects that manifest only under certain stress conditions or timepoints
While direct evidence for Samd3's role in cell cycle regulation is limited, insights can be drawn from related proteins like SAMD5, which has been implicated in cell cycle regulation of cholangiocarcinoma cell lines . By extension, researchers might investigate Samd3's potential role through:
Experimental Approaches:
siRNA knockdown in relevant cell lines followed by cell cycle analysis
Overexpression studies to assess effects on proliferation
ChIP-seq to identify potential binding to cell cycle regulatory genes
Co-immunoprecipitation to identify interaction partners
Cell Cycle Analysis Methods:
Flow cytometry with propidium iodide or DAPI for DNA content
BrdU incorporation assays to measure S-phase entry
Live cell imaging with fluorescent cell cycle reporters
Western blotting for cyclins and CDK inhibitors
Based on findings with SAMD5, where knockdown accelerated proliferation and increased S and G2/M phase populations , researchers might hypothesize that Samd3 could exert similar regulatory effects on lymphocyte proliferation.
The current literature presents an interesting contradiction: Samd3 expression is enriched in memory precursor cells and exhausted T cell precursors, suggesting functional importance, yet Samd3-deficient CD8+ T cells were not compromised in viral infection models . To address such contradictions:
Methodological Approach:
Implement rigorous experimental controls including genetic background matching
Use multiple infection models with varying pathogen doses and routes
Extend observation periods for long-term memory or exhaustion phenotypes
Apply single-cell analysis to detect subtle phenotypes in specific subpopulations
Data Interpretation Framework:
Consider redundancy with other SAM-domain proteins
Evaluate compensatory mechanisms that may mask phenotypes
Assess environmental factors that might influence gene-function relationships
Analyze strain-specific effects that could affect reproducibility
Resolution Strategies:
Develop double or triple knockout models to address redundancy
Use competitive adoptive transfer experiments to enhance sensitivity
Implement stress conditions that might reveal conditional phenotypes
Consider tissue-specific functions that might not be apparent in systemic analyses
When designing experiments to study Samd3 in immune responses, researchers should carefully consider the balance between abstraction and detail in their experimental approach:
Abstraction vs. Detail Considerations:
Experimental System Selection:
In vitro: Cell lines provide control but lack physiological context
Ex vivo: Primary cells maintain more native properties but introduce variability
In vivo: Animal models offer systemic context but increase complexity
Each approach has strengths and limitations that should be explicitly acknowledged
Construct Validity Considerations:
Data Integration Table:
| Approach | Strengths | Limitations | Best For |
|---|---|---|---|
| Cell Lines | High control, low variability | Limited physiological relevance | Mechanism studies, protein interactions |
| Primary Cells | Physiologically relevant | Donor variability | Ex vivo functional assays, signaling studies |
| Reporter Mouse | Visualize expression dynamics | Potential reporter interference | Expression patterns, cell tracking |
| Conditional KO | Temporal and spatial specificity | Incomplete recombination | Cell-specific functions, developmental roles |
| Global KO | Complete gene deletion | Developmental compensation | Essential functions, redundancy assessment |