The protein is commercially available in multiple recombinant forms:
Full-length isoform 2: Includes residues 1–80, optimized for structural studies .
Partial constructs: Truncated versions for specific antibody generation or binding assays .
Membrane Protein Dynamics: Used to investigate the role of SMIM4 in lipid bilayer interactions due to its integral membrane nature .
Gene Editing: While not directly linked to CRISPR/Cas9 systems, recombinant UPF0640 homologs are employed in parallel studies involving RAD51-enhanced homologous recombination (e.g., optimizing knock-in efficiency in mouse embryos) .
Mass Spectrometry: Validates molecular weight and sequence integrity .
Functional Assays: Binding studies with putative interactors (e.g., lipid membranes) .
Condition | Stability |
---|---|
Short-term Storage | 1 month at 4°C |
Long-term Storage | >12 months at -80°C |
Reconstitution | 100 μg/mL in PBS; avoid vortexing |
UPF0640 protein C3orf78 homolog is classified as a small integral membrane protein that is also referred to as small integral membrane protein 4 in some databases . While the complete functional characterization remains under investigation, it belongs to the ubiquinol-cytochrome c reductase complex assembly factor family, suggesting a potential role in mitochondrial electron transport chain assembly and function. As a membrane protein, it likely participates in cellular signaling pathways or transport mechanisms across cellular compartments.
The mouse homolog shares significant sequence similarity with the human UPF0640 protein C3orf78, though species-specific variations in protein structure and function may exist. Current research approaches typically involve comparative genomic analysis between mouse and human variants to elucidate conserved functional domains and potential physiological roles.
The structural characteristics of mouse UPF0640 protein C3orf78 homolog differ significantly from well-characterized mouse recombinant proteins such as OB protein and ULBP-1/MULT-1. Unlike OB protein, which has been extensively studied for its role in body weight regulation , UPF0640 protein C3orf78 homolog has a membrane-associated domain architecture. The protein likely contains transmembrane helices that anchor it to cellular membranes, which presents unique challenges for expression and purification compared to soluble proteins like recombinant mouse monoclonal antibodies .
Similar to other membrane proteins, the structural integrity of UPF0640 protein C3orf78 homolog depends heavily on proper post-translational modifications and membrane environment. When designing experimental approaches, researchers should consider that, unlike the mouse ULBP-1/MULT-1 protein which has well-defined alpha-1 and alpha-2 domains with intrachain disulfide bonds , the structural elements of UPF0640 protein C3orf78 homolog may require specialized techniques for proper folding and stabilization during recombinant expression.
The choice of expression system for recombinant mouse UPF0640 protein C3orf78 homolog depends on research objectives and required protein characteristics. Multiple expression platforms offer distinct advantages:
Expression System | Advantages | Disadvantages | Best For |
---|---|---|---|
E. coli | High yield, rapid production, cost-effective, simpler purification protocols | Limited post-translational modifications, potential inclusion body formation | Structural studies requiring large quantities, preliminary characterization |
Yeast (P. pastoris, S. cerevisiae) | Moderate to high yield, eukaryotic post-translational modifications, secretion capabilities | Longer production time than bacteria, hyperglycosylation issues | Functional studies requiring some post-translational modifications |
Insect cells with baculovirus | Complex eukaryotic post-translational modifications, good for membrane proteins | Higher cost, longer production timeline, technical complexity | Studies requiring authentic folding and more complex modifications |
Mammalian cells | Most physiologically relevant post-translational modifications, native-like protein folding | Highest cost, lowest yield, longest timeline | Studies requiring complete biological activity and mammalian-specific modifications |
Purification of recombinant mouse UPF0640 protein C3orf78 homolog requires specialized approaches due to its membrane protein nature. Effective purification typically involves a multi-step strategy:
Membrane extraction: Utilizing detergents like n-dodecyl-β-D-maltoside (DDM), CHAPS, or digitonin to solubilize the protein from cellular membranes while maintaining structural integrity.
Affinity chromatography: Expression with fusion tags (His-tag, similar to the approach used with mouse ULBP-1/MULT-1 protein ) enables selective capture on affinity resins. His-tagged variants can be purified using immobilized metal affinity chromatography (IMAC).
Size exclusion chromatography: Further purification based on molecular size helps separate the target protein from aggregates and other cellular components.
Ion exchange chromatography: Additional purification step based on the protein's charge characteristics at specific pH values.
For membrane proteins like UPF0640, maintaining the protein in a detergent micelle or reconstituting it into lipid nanodiscs or liposomes post-purification is often necessary to preserve native structure and function. Researchers should monitor protein stability throughout the purification process using techniques such as dynamic light scattering or size exclusion chromatography with multi-angle light scattering (SEC-MALS).
Optimizing stability and solubility of recombinant mouse UPF0640 protein C3orf78 homolog requires careful consideration of several factors:
Expression temperature: Lower temperatures (16-20°C) often improve proper folding and reduce aggregation, particularly important for membrane proteins.
Induction conditions: For bacteria-based systems, optimizing IPTG concentration and induction timing can significantly impact protein folding and yield.
Fusion partners: Solubility-enhancing fusion tags like MBP (maltose-binding protein), GST (glutathione S-transferase), or SUMO can improve expression of membrane proteins.
Buffer composition: Screening different buffers, pH conditions, and salt concentrations is essential for maintaining protein stability during purification.
Additives and stabilizers: Addition of glycerol (10-15%), specific lipids, or mild detergents can enhance stability.
Carrier proteins: Similar to the approach used with recombinant mouse ULBP-1/MULT-1 protein, adding carrier proteins like BSA can enhance protein stability and increase shelf-life .
A systematic approach using multivariate experimental designs, such as the Box-Behnken design illustrated in search result , allows efficient optimization of multiple parameters simultaneously. These designs are particularly valuable when working with limited quantities of protein and enable identification of optimal conditions that might not be discovered through conventional approaches where only one parameter is varied at a time .
Effective optimization of recombinant mouse UPF0640 protein C3orf78 homolog production benefits from systematic multivariate experimental design approaches rather than one-factor-at-a-time methods. Key experimental design strategies include:
Central Composite Design: This three-dimensional (or higher) design incorporates center points (all variables at mid-levels), axial points (one variable at high/low level, others at mid-level), and factorial points (all variables at high/low levels) . This approach is particularly useful for identifying optimal expression conditions when multiple parameters interact.
Box-Behnken Design: This efficient design varies parameters two at a time while keeping others at their mid-levels . It's particularly useful when extreme combinations of all factors might lead to experimental failures (such as protein aggregation at both high temperature and high concentrations).
Full Factorial Design: While resource-intensive, this approach tests all possible combinations of parameters and is most comprehensive for identifying complex interactions.
For recombinant mouse UPF0640 protein C3orf78 homolog expression, key parameters to optimize through these designs include:
Protein concentration
Inducer concentration
Temperature
pH
Media composition
Harvest time
As demonstrated in the thaumatin crystallization example, multivariate designs can reveal optimal conditions that would not be discovered in conventional approaches . For instance, varying protein concentration—a parameter not typically changed in first-round optimization—may yield significantly better results, as shown in Figure 6 of source .
Comprehensive functional characterization of recombinant mouse UPF0640 protein C3orf78 homolog requires multiple complementary approaches:
Binding assays: Surface plasmon resonance (SPR) or microscale thermophoresis (MST) can determine binding affinities with potential interaction partners.
Activity assays: If the protein is involved in electron transport as suggested by its classification, oxygen consumption rate measurements or enzyme-coupled assays may be appropriate.
Localization studies: Fluorescently tagged versions can be used to determine subcellular localization in live cells.
Knockout/knockdown studies: CRISPR-Cas9 mediated gene editing or siRNA approaches to assess phenotypic consequences of protein absence.
Structural studies: Cryo-EM or X-ray crystallography can provide structural insights, though membrane proteins present unique challenges.
Protein-protein interaction mapping: Co-immunoprecipitation followed by mass spectrometry or yeast two-hybrid screens can identify interaction partners.
Reconstitution experiments: For functional validation, purified protein can be reconstituted into liposomes or nanodiscs to assess transport or enzymatic activities.
These approaches should be applied systematically, beginning with localization and interaction studies to generate hypotheses about function, followed by more targeted biochemical assays based on preliminary findings.
Comprehensive quality control for recombinant mouse UPF0640 protein C3orf78 homolog should include multiple analytical techniques:
SDS-PAGE: Basic assessment of purity and approximate molecular weight, with expected band corresponding to the theoretical molecular weight plus any fusion tags.
Western blotting: Verification of protein identity using antibodies against the protein itself or common epitope tags (His, FLAG, etc.).
Mass spectrometry:
MALDI-TOF or ESI-MS for accurate molecular weight determination
Peptide mapping through tryptic digestion and LC-MS/MS for sequence coverage and identification of post-translational modifications
Circular dichroism (CD): Assessment of secondary structure to confirm proper folding.
Size exclusion chromatography (SEC): Evaluation of oligomeric state and detection of aggregates.
Dynamic light scattering (DLS): Measurement of particle size distribution to assess homogeneity.
Endotoxin testing: Particularly important for proteins intended for cell-based assays.
For membrane proteins like UPF0640, additional quality control measures should include detergent content analysis and assessment of lipid composition if the protein is reconstituted into liposomes or nanodiscs.
Determining the half-life and stability profile of recombinant mouse UPF0640 protein C3orf78 homolog requires multiple analytical approaches:
In vitro stability studies:
Thermal stability using differential scanning fluorimetry (DSF) or differential scanning calorimetry (DSC)
Storage stability at different temperatures (4°C, -20°C, -80°C)
Freeze-thaw stability through multiple cycles
pH stability across physiologically relevant range
In vivo half-life determination:
Similar to approaches used for mouse monoclonal antibodies, which involve intravenous administration followed by serial sampling to determine serum concentrations over time
Analysis typically reveals biphasic kinetics with distinct alpha (distribution) and beta (elimination) phases
Clearance mechanisms may differ from antibodies due to the membrane protein nature
Accelerated stability studies:
Exposure to elevated temperatures to predict long-term stability
Analysis by SEC, SDS-PAGE, and functional assays at defined time points
When developing storage protocols, researchers should consider that membrane proteins often require specialized stabilization strategies similar to those used for mouse ULBP-1/MULT-1 protein, such as lyophilization from filtered PBS solutions and storage in manual defrost freezers to avoid freeze-thaw cycles .
Expressing recombinant mouse UPF0640 protein C3orf78 homolog presents several challenges common to membrane proteins:
Challenge | Causes | Solutions |
---|---|---|
Low expression levels | Toxicity to host cells, inefficient translation, poor membrane insertion | Use tightly regulated inducible promoters, specialized host strains (C41/C43 for E. coli), codon optimization, fusion partners |
Protein misfolding | Improper membrane insertion, absence of chaperones, rapid expression | Reduce expression temperature, co-express with chaperones, use specialized folding-promoting additives |
Aggregation during purification | Detergent-mediated destabilization, removal from native membrane environment | Screen multiple detergents, use amphipols or nanodiscs, include stabilizing lipids |
Loss of function | Denaturation during purification, critical post-translational modifications missing | Use activity assays throughout purification, select expression system providing necessary modifications |
Proteolytic degradation | Exposure to host proteases, intrinsic instability | Include protease inhibitors, engineer more stable variants, reduce purification time |
A methodical troubleshooting approach involves systematically varying expression conditions through multivariate experimental designs like those described in source . For recombinant mouse UPF0640 protein, particular attention should be paid to membrane mimetics during purification and reconstitution to maintain native-like environment.
Contradictory results in functional studies of recombinant mouse UPF0640 protein C3orf78 homolog may arise from several factors, and reconciling these discrepancies requires a systematic approach:
Expression system differences: Variations in post-translational modifications between bacterial, yeast, insect, and mammalian expression systems can significantly impact protein function . Researchers should directly compare protein from different sources using consistent functional assays.
Protein preparation variables: Different purification methods, detergents, or reconstitution systems may preserve different functional aspects. Comprehensive characterization using multiple biochemical and biophysical techniques can identify preparation-dependent differences.
Experimental design inconsistencies: Variations in buffer conditions, temperature, pH, or presence of cofactors can affect results. Applying systematic multivariate experimental designs similar to those described for crystallization optimization can help identify critical parameters affecting function.
Tag interference: Fusion tags may interfere with certain functions while preserving others. Comparing tagged and untagged versions, or versions with tags at different positions (N-terminal vs. C-terminal), can resolve tag-dependent effects.
Binding partner variations: If contradictory results involve protein-protein interactions, differences may reflect true biological variability in binding affinities depending on cellular context or post-translational modification state.
When faced with contradictory results, researchers should consider replicating experiments with protein prepared using multiple methods while systematically controlling variables to identify the source of discrepancies.
Emerging research applications for recombinant mouse UPF0640 protein C3orf78 homolog span multiple areas of biomedical research:
Structural biology advances: Cryo-EM approaches optimized for membrane proteins may reveal the first high-resolution structures of this protein family, potentially illuminating functional mechanisms.
Drug discovery applications: As a potential component of mitochondrial respiration pathways, the protein may represent a novel target for metabolic disorders or mitochondrial diseases. Recombinant protein enables development of high-throughput screening assays.
Comparative biology: Systematic comparison between mouse and human homologs can provide evolutionary insights into conserved functions and species-specific adaptations.
Synthetic biology: Integration into artificial membrane systems or engineered cellular pathways may enable novel biosensing or bioenergetic applications.
Systems biology: Network analysis incorporating interaction partners identified through proteomics approaches may reveal unexpected cellular roles beyond currently predicted functions.
CRISPR-mediated functional genomics: Combining recombinant protein studies with gene editing approaches enables validation of function in cellular and animal models.
These emerging applications highlight the importance of developing robust expression and purification protocols that yield protein with native-like characteristics. Researchers should consider collaborative approaches that leverage complementary expertise in structural biology, functional genomics, and computational modeling.