Recombinant Mouse Uncharacterized protein C17orf109 homolog

Shipped with Ice Packs
In Stock

Description

Definition and Homology

The Recombinant Mouse Uncharacterized Protein C17orf109 Homolog refers to a synthetic version of the mouse homolog of the human gene C17orf109. This gene encodes a small integral membrane protein (SMIM5) that remains functionally uncharacterized in both humans and mice . Homology analysis reveals conservation across species, with SMIM5 being the human ortholog and Smim5 the mouse counterpart .

Gene and Protein Characteristics

FeatureDetails
Gene SymbolSmim5 (mouse), SMIM5 (human)
Protein LengthPredicted full-length sequence (exact length varies by species)
Membrane AssociationIntegral membrane protein (predicted transmembrane domains)
ExpressionWidely expressed in tissues, though specific roles are undefined
HomologsC17orf109 (human), Smim5 (mouse), Smim5 (rat)

The protein’s structure includes hydrophobic regions consistent with transmembrane domains, suggesting a role in membrane-bound processes .

Challenges and Future Directions

  • Functional Elucidation: The lack of catalytic domains in Smim5 suggests a regulatory or structural role, requiring further biochemical assays .

  • Homology-Based Hypotheses: Comparative studies with ATP synthase subunits (e.g., subunit f) may reveal evolutionary conserved functions .

  • Therapeutic Potential: While speculative, uncharacterized membrane proteins are often targets in disease research (e.g., mitochondrial disorders) .

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 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 advance notification 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 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 forms have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is essential for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during the manufacturing process.
The specific tag will be determined during production. If you require a specific tag, please inform us, and we will prioritize its development.
Synonyms
Smim5; Small integral membrane protein 5
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-78
Protein Length
full length protein
Species
Mus musculus (Mouse)
Target Names
Smim5
Target Protein Sequence
MAATDFVGEIRSVGERLLLKLQQLPQAEPVELVAFSIIVLFTATVLVLGLIACSCCCAHC CCSESRQRKIPVRPTKPR
Uniprot No.

Target Background

Database Links

KEGG: mmu:66528

UniGene: Mm.275740

Subcellular Location
Membrane; Single-pass membrane protein.

Q&A

What is Recombinant Mouse Uncharacterized protein C17orf109 homolog?

Recombinant Mouse Uncharacterized protein C17orf109 homolog is a protein of interest in research settings that has not yet been fully characterized functionally. It belongs to a class of proteins initially identified through genomic sequencing but whose biological functions have not been completely elucidated. Similar to other uncharacterized proteins such as IL-34 (formerly known as uncharacterized protein C16orf77 homolog), this protein may eventually be assigned a specific name and function following comprehensive research and characterization .

What expression systems are most suitable for producing Recombinant Mouse Uncharacterized protein C17orf109 homolog?

Multiple expression systems can be utilized for the production of Recombinant Mouse Uncharacterized protein C17orf109 homolog, each with distinct advantages. Bacterial systems (E. coli) and yeast typically offer superior yields and shorter production timeframes, making them cost-effective options for initial studies. Insect cell expression using baculovirus and mammalian cell expression systems provide more complex post-translational modifications that may be critical for proper protein folding and biological activity maintenance . The optimal choice depends on your specific research requirements regarding protein authenticity, yield requirements, and functional activity needs.

How does protein purification strategy affect downstream experimental applications?

The purification strategy significantly impacts protein quality and subsequent experimental outcomes. For Recombinant Mouse Uncharacterized protein C17orf109 homolog, a multi-step chromatographic approach similar to that used for other recombinant proteins is recommended. Typically, this includes an initial capture step using affinity chromatography (if the protein is tagged) or ion exchange chromatography, followed by polishing steps using size-exclusion chromatography to achieve high purity levels . The final purity should be assessed using analytical techniques such as SEC-HPLC, with target purity levels exceeding 95% to ensure reliable experimental results . Different downstream applications may require specific buffer formulations and protein concentrations to maintain stability and activity.

What are the optimal expression conditions for maximizing protein yield in E. coli systems?

For E. coli expression systems, optimizing induction parameters is critical for maximizing yield of Recombinant Mouse Uncharacterized protein C17orf109 homolog. Temperature, inducer concentration, and induction timing significantly impact protein expression levels. Lower induction temperatures (16-25°C) often improve soluble protein yields by reducing inclusion body formation. For IPTG-inducible systems, concentrations between 0.1-1.0 mM and induction at mid-log phase (OD600 of 0.6-0.8) frequently yield optimal results. Additionally, supplement the culture medium with specific amino acids or cofactors if the protein contains structural elements requiring them. Expression should be monitored using SDS-PAGE analysis at multiple time points to determine the optimal harvest time, typically 4-24 hours post-induction depending on protein characteristics and expression system parameters.

How can mammalian expression systems be optimized for producing properly folded Recombinant Mouse Uncharacterized protein C17orf109 homolog?

Mammalian expression systems, particularly Chinese Hamster Ovary (CHO) cells, offer significant advantages for producing properly folded complex proteins with appropriate post-translational modifications. Establishing stable transfectants requires a systematic selection strategy using appropriate selection markers. A two-phase selection scheme employing different concentrations of selection agents (such as methotrexate and puromycin) can effectively isolate high-producing clones . For CHO cell cultures, optimization of culture parameters including temperature (typically 37°C initially, with potential reduction to 30-33°C during production phase), pH (6.8-7.2), dissolved oxygen (30-60%), and feeding strategy significantly impacts protein yield and quality. Cell culture medium formulation should be optimized to support both cell growth and protein production, with potential supplements including various growth factors and nutrient mixtures.

What purification techniques yield the highest recovery of active protein?

A multi-step purification strategy typically yields the highest recovery of active Recombinant Mouse Uncharacterized protein C17orf109 homolog. An effective approach involves initial capture using ion exchange chromatography (typically cation exchange for most proteins with pI > 7) followed by size-exclusion chromatography for final polishing . This combination can achieve purity levels exceeding 98% while maintaining biological activity. Throughout the purification process, buffer conditions (pH, ionic strength, stabilizing additives) should be optimized to maintain protein stability and activity. The purification should be validated using analytical techniques including SEC-HPLC, which can quantitatively assess purity, and functional assays to confirm biological activity is preserved . Recovery rates typically range from 50-80% of the total expressed protein, with functional activity retention being the critical quality attribute.

What bioassays are appropriate for confirming the biological activity of Recombinant Mouse Uncharacterized protein C17orf109 homolog?

The selection of appropriate bioassays depends on the predicted functional characteristics of the protein based on sequence homology and structural features. If the protein shares homology with cytokines like IL-34, cell-based proliferation assays using responsive cell lines would be appropriate. For example, TF-1 cells are commonly used for assessing cytokine activity, with activity measured using MTS/MTT assays or direct cell counting . The specific activity determination requires dose-response experiments, typically performed in triplicate with appropriate positive controls. For initial characterization, comparing the activity profile with that of similar characterized proteins can provide valuable insights into potential functions. The ED50 values (effective dose for 50% maximal response) should be calculated and reported in ng/mL to enable comparison with established standards.

How can researchers distinguish between native biological activity and artificial effects in functional assays?

To distinguish between true biological activity and artificial effects, implement rigorous controls in all functional assays. Include appropriate negative controls (inactive protein versions, buffer-only treatments), positive controls (well-characterized related proteins), and specificity controls (competing ligands or receptor-blocking antibodies when receptors are known). Statistical validation across multiple independent experiments is essential. Additionally, corroborate results using complementary assay methodologies measuring different aspects of the same biological effect. For instance, if studying potential effects on cell proliferation, combine metabolic activity assays (MTS/MTT) with direct cell counting and cell cycle analysis. Consider using primary cells in addition to established cell lines to verify the physiological relevance of observed effects. Dose-response relationships should follow expected patterns for receptor-mediated biological effects (typically sigmoidal curves with plateaus at high concentrations).

What structural analyses should be performed to confirm proper folding of the recombinant protein?

Multiple complementary analytical techniques should be employed to comprehensively assess the structural integrity of Recombinant Mouse Uncharacterized protein C17orf109 homolog. These include:

  • Circular dichroism (CD) spectroscopy to evaluate secondary structural elements

  • Intrinsic fluorescence analysis to assess tertiary structure

  • Size-exclusion chromatography to detect aggregation states

  • Differential scanning calorimetry to determine thermal stability

  • N-terminal sequencing to confirm protein identity and processing

  • Mass spectrometry for accurate molecular weight determination and post-translational modification analysis

The collective data from these analyses should be compared with theoretical predictions based on sequence analysis and homology to related proteins with known structures. Proper folding is ultimately validated by demonstrating biological activity in functional assays, as misfolded proteins typically show reduced or absent activity.

How should researchers design experiments to investigate potential binding partners of Uncharacterized protein C17orf109 homolog?

A systematic approach to identifying binding partners involves multiple complementary techniques:

  • Affinity Purification coupled with Mass Spectrometry (AP-MS): Express the recombinant protein with an affinity tag, use it as bait to capture interacting proteins from cell lysates, and identify binding partners through mass spectrometry.

  • Yeast Two-Hybrid Screening: Useful for detecting direct protein-protein interactions, though prone to false positives requiring validation.

  • Surface Plasmon Resonance (SPR): Provides quantitative binding parameters (association/dissociation constants) with candidate binding partners.

  • Co-immunoprecipitation: Validates interactions in more physiologically relevant contexts using specific antibodies.

For experimental design, follow established guidelines for biological replication (minimum 3 independent experiments) and include appropriate controls (non-specific proteins of similar size/charge, mutant versions of the protein) . Consider conducting experiments in multiple cell types or tissues to identify context-specific interactions. Validation of key interactions should employ at least two independent methodologies.

What controls are essential when investigating the effects of Recombinant Mouse Uncharacterized protein C17orf109 homolog on cellular processes?

Essential controls for investigations of cellular effects include:

  • Vehicle Controls: Buffer-only treatments that match the recombinant protein formulation.

  • Concentration Series: Multiple concentrations to establish dose-response relationships.

  • Heat-inactivated Protein: To distinguish between effects requiring native protein structure versus non-specific effects.

  • Endotoxin Controls: Especially for E. coli-expressed proteins, to rule out LPS contamination effects.

  • Related Protein Controls: Structurally similar proteins to establish specificity of observed effects.

  • Blocking Controls: If potential receptors are known, include receptor-blocking antibodies or competitive ligands.

The experimental design should incorporate randomization, blinding where possible, and appropriate statistical considerations for sample size determination . Time-course studies are also valuable for distinguishing between direct effects and secondary consequences of protein treatment.

How can researchers optimize storage conditions to maintain stability of purified Recombinant Mouse Uncharacterized protein C17orf109 homolog?

Optimal storage conditions should be systematically determined through stability studies. For lyophilized preparations, storage at -20°C or -80°C in a manual defrost freezer is typically recommended to avoid freeze-thaw cycles . For reconstituted protein, stability should be assessed in different buffer formulations, with and without stabilizing additives such as bovine serum albumin (BSA) .

Aliquoting is essential to avoid repeated freeze-thaw cycles, which can significantly reduce protein activity. Stability should be monitored using both structural (SEC-HPLC, CD spectroscopy) and functional assays at defined time points (initial, 1 month, 3 months, 6 months, 1 year) under different storage conditions.

For carrier-free preparations intended for applications where BSA might interfere, special consideration should be given to buffer optimization, potentially incorporating alternative stabilizers such as sugars (trehalose, sucrose) or specific amino acids . Record and report protein stability data using standardized metrics such as percent activity retention over time.

What are the appropriate methodologies for investigating potential receptor interactions of Uncharacterized protein C17orf109 homolog?

Investigation of potential receptor interactions requires a multi-faceted approach:

  • Receptor Binding Assays: Using radiolabeled or fluorescently labeled purified protein to quantify binding to cell surfaces, followed by competition assays with unlabeled protein to determine specificity.

  • Phosphorylation Studies: If the protein is suspected to be a ligand like IL-34, investigate activation of signaling pathways by assessing phosphorylation of molecules like ERK1/2 .

  • CRISPR/Cas9 Receptor Knockout: Generate receptor-knockout cell lines to confirm specific receptor dependencies.

  • Receptor Cross-linking: Chemical cross-linking followed by immunoprecipitation and mass spectrometry to identify unknown receptors.

  • Functional Reporter Assays: Develop cell lines expressing potential receptors coupled to reporter systems (luciferase, GFP) responding to receptor activation.

Correlation of receptor binding parameters with functional activity is essential for establishing physiologically relevant interactions. Compare binding characteristics with those of related proteins that have well-characterized receptor interactions to identify similarities and differences in receptor recognition patterns.

How can researchers develop cell-based assays to investigate the biological function of Uncharacterized protein C17orf109 homolog?

Development of cell-based assays should proceed through systematic steps:

  • Cell Type Selection: Based on tissues with high expression of the protein or its homologs, or cells known to respond to functionally similar proteins. For instance, if the protein shares homology with IL-34, monocytes or macrophage progenitors would be appropriate .

  • Endpoint Selection: Choose biologically relevant endpoints such as cell viability, proliferation, differentiation, or specific gene expression changes. For cytokine-like proteins, colony formation assays may be informative .

  • Assay Optimization: Systematically optimize cell density, protein concentration range, incubation time, and detection methods.

  • Validation: Confirm assay reproducibility through statistical analysis of multiple independent experiments, and establish Z-factor values for high-throughput applications.

  • Mechanistic Studies: Use inhibitors of specific signaling pathways to elucidate the mechanisms underlying observed effects.

Development of a standardized positive control (benchmark protein with similar function) is valuable for normalizing results across experiments and laboratories.

What approaches are recommended for investigating the in vivo functions of Uncharacterized protein C17orf109 homolog?

In vivo functional investigation requires:

  • Expression Analysis: Comprehensive tissue distribution profiling using qRT-PCR, western blotting, and immunohistochemistry to identify tissues with significant expression .

  • Genetic Models: Development of knockout and/or transgenic overexpression mouse models to study loss-of-function and gain-of-function phenotypes.

  • Administration Studies: Direct administration of purified recombinant protein to wild-type animals with comprehensive phenotypic analysis.

  • Developmental Analysis: Investigation of temporal expression patterns during embryonic and postnatal development if developmentally regulated functions are suspected.

  • Disease Models: Testing effects in relevant disease models based on expression patterns and in vitro functional data.

Systematic phenotyping should include analysis of all major physiological systems, with particular focus on tissues with high expression levels. For proteins with potential roles in specific cellular processes (e.g., differentiation, proliferation), appropriate cellular analyses of affected tissues should be performed. Correlate in vivo findings with in vitro mechanistic studies to develop a comprehensive understanding of biological function.

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.