Recombinant Human Uncharacterized protein UNQ511/PRO1026, also referred to as UNQ511/PRO1026, is a protein whose function has not been fully elucidated. It is part of a broader category of uncharacterized proteins, which are proteins that have been identified through genomic sequencing but have not yet been studied extensively in terms of their biological roles or functions. This protein is often associated with the gene LYPD8, which encodes a ly6/PLAUR domain-containing protein 8 .
Gene Name: LYPD8
Protein Name: Ly6/PLAUR domain-containing protein 8
Host/Reactivities: The recombinant protein can be expressed in various hosts such as E. coli, yeast, baculovirus, or mammalian cells .
Purity: Typically, the purity of recombinant proteins is greater than or equal to 85% as determined by SDS-PAGE .
Recombinant proteins like UNQ511/PRO1026 are produced through recombinant DNA technology, where the gene encoding the protein is inserted into a suitable expression vector and then expressed in a host organism. The choice of host depends on the desired level of post-translational modification and the ease of purification. For instance, mammalian cells can provide more complex post-translational modifications compared to bacterial systems.
While the specific biological function of UNQ511/PRO1026 remains uncharacterized, proteins within the ly6/PLAUR domain family are generally involved in cell surface interactions and may play roles in immune responses or cellular adhesion processes. Further research is needed to elucidate its exact role in human biology.
| Characteristic | Description |
|---|---|
| Gene Name | LYPD8 |
| Protein Name | Ly6/PLAUR domain-containing protein 8 |
| Host/Reactivities | E. coli, Yeast, Baculovirus, Mammalian Cells |
| Purity | ≥85% (SDS-PAGE) |
| Expression Region | Variable depending on construct |
| Potential Biological Role | Cell surface interactions, immune responses |
Future studies on UNQ511/PRO1026 could involve functional assays to determine its role in cellular processes. Techniques such as co-immunoprecipitation to identify interacting proteins, or RNA interference to assess its impact on cell behavior, could provide valuable insights. Additionally, structural studies could help elucidate how its ly6/PLAUR domain contributes to its function.
This secreted protein plays a crucial role in preventing Gram-negative bacterial invasion of the colon's inner mucus layer, a commensal microbiota-free region of the large intestine. It inhibits bacterial motility by binding to bacterial flagella (e.g., P. mirabilis), thereby maintaining intestinal homeostasis through the spatial separation of intestinal bacteria and epithelial cells.
For successful recombinant expression of uncharacterized proteins like UNQ511/PRO1026, several methodological considerations are critical. First, sequence optimization is essential - this includes altering suboptimal codon usage for mammalian tRNA bias, improving secondary mRNA structure, and removing AT-rich regions to increase mRNA stability . For expression, it's recommended to generate a construct without the transmembrane domain (if present) to improve solubility, as demonstrated in successful recombinant protein studies .
Expression systems should be selected based on the protein's predicted properties. For uncharacterized human proteins:
Mammalian expression systems (HEK293 or CHO cells) maintain proper folding and post-translational modifications
Bacterial systems (E. coli) are suitable for proteins without complex folding requirements
Insect cell systems (Sf9, Hi5) offer a compromise between yield and proper folding
For purification, adding a C-terminal tag (such as a 10-His tag) facilitates isolation while minimizing interference with protein function . After expression, reconstitution at 100 μg/mL in sterile PBS is typically suitable for initial characterization studies .
Initial characterization of uncharacterized proteins should follow a systematic workflow. Begin with bioinformatic analysis to predict domains, motifs, and potential functions based on sequence homology. Next, confirm protein expression through Western blotting and mass spectrometry, requiring at least two detected peptides (at least one unique) with FDR cut-off set to 1% .
For biochemical characterization, analyze:
Protein stability under different conditions (pH, temperature, buffer compositions)
Secondary structure elements using circular dichroism
Post-translational modifications using mass spectrometry
Oligomerization state using size exclusion chromatography
Most critically, perform protein-protein interaction (PPI) analysis using affinity purification-mass spectrometry (AP-MS) to identify binding partners . This approach has successfully predicted Gene Ontology categories for 387 previously uncharacterized proteins . When conducting these experiments, ensure proper controls including unrelated proteins with similar biochemical properties to distinguish specific from non-specific interactions.
Quality control for recombinant uncharacterized proteins requires rigorous methodology. Purity assessment should employ multiple techniques including SDS-PAGE (>95% purity), mass spectrometry to confirm molecular weight and absence of major contaminants, and dynamic light scattering to assess homogeneity .
For functional integrity assessment:
Verify proper folding using circular dichroism or fluorescence spectroscopy
Confirm biological activity through preliminary binding assays with predicted partners
Assess aggregation propensity during storage using size exclusion chromatography
Stability testing is essential - lyophilized protein formulations should be assessed for activity retention after reconstitution, and repeated freeze-thaw cycles should be avoided as they can compromise protein integrity . For storage, use a manual defrost freezer and aliquot the protein to minimize freeze-thaw cycles . Documentation should include detailed records of expression conditions, purification methods, batch-to-batch variability, and storage conditions to ensure reproducibility across experiments.
Protein-protein interaction (PPI) networks provide powerful insights into functions of uncharacterized proteins. For UNQ511/PRO1026, researchers should construct comprehensive PPI networks using AP-MS experiments similar to those employed in BioPlex 2.0 . This approach involves tagging the protein of interest (HA-FLAG-tagged open reading frames), expressing it in appropriate cell lines, and identifying interaction partners through mass spectrometry .
The resulting network should be analyzed using multiple computational approaches:
Guilt-by-association analysis - assigning functions based on known functions of interaction partners
Network topology analysis - identifying whether the protein functions as a hub or peripheral component
Clustering coefficient calculation - determining if the protein participates in functional modules
Betweenness centrality measurement - assessing the protein's role in connecting different cellular processes
In a study of uncharacterized proteins, researchers constructed a PPI network with 9,967 vertices connected by 287,474 interactions, with 8,686 proteins forming a single giant component . Each protein interacted with approximately 5 partners (median value) . By applying similar methodology to UNQ511/PRO1026, researchers can predict cellular compartmentalization, molecular function, and biological processes with statistical confidence.
Examine control group selection - different reference points may yield different interpretations
Evaluate expression system influences - protein function may vary between mammalian, bacterial, or cell-free systems
Assess tag interference - different fusion tags may alter protein behavior
Consider post-translational modification variations across experimental conditions
Analyze splice form differences - alternative splicing may yield functionally distinct proteoforms
When designing resolution experiments, employ factorial experimental design to systematically test multiple variables simultaneously. For example, in BioPlex studies, researchers identified that 27 genes encoding uPE1 proteins were detected in both canonical and splice forms, potentially explaining functional differences . Similarly, for UNQ511/PRO1026, examining both canonical and alternative splice forms is critical for comprehensive characterization and resolving contradictory findings.
Alternative splicing can generate functionally distinct proteoforms of uncharacterized proteins. To identify such differences in UNQ511/PRO1026, researchers should utilize a multi-faceted approach. First, employ RNA sequencing to identify all expressed splice variants in relevant tissues and quantify their relative abundance . Next, design expression constructs for each splice variant with identical tags to ensure comparative analysis.
For functional comparison between variants:
Perform comparative interactome analysis - as demonstrated in BioPlex 2.0, where functional differences were revealed for 62 proteoforms encoded by 31 genes
Conduct domain-specific assays based on predicted structural differences
Utilize subcellular localization studies to identify differential compartmentalization
Implement CRISPR-based isoform-specific knockdown to assess variant-specific phenotypes
When analyzing interaction data, researchers should construct separate PPI networks for each splice variant. In previous studies, researchers detected distinct functional profiles for canonical and alternatively spliced forms for four uPE1 genes . This approach can reveal whether different UNQ511/PRO1026 variants participate in distinct cellular processes or interact with different partner proteins, providing critical insights into their specialized functions.
If UNQ511/PRO1026 is suspected to participate in signaling pathways, kinetic modulation assays can provide functional insights. These assays assess how the protein affects the binding kinetics and signaling dynamics of known biological complexes. Researchers should develop polypeptide binding agents (e.g., antibodies) that can modulate the protein's activity and assess consequent effects on signaling .
A comprehensive kinetic modulation approach includes:
Generation of binding agents - use phage display technology to develop human monoclonal antibodies against the target protein
Binding kinetics assessment - employ surface plasmon resonance to measure association and dissociation rates
Signaling assays - develop cell-based reporter systems to measure effects on downstream signaling pathways
Dose-response relationships - establish quantitative relationships between binding agent concentration and signaling outcomes
These kinetic modulators can function as modulators, potentiators, regulators, effectors or inhibitors depending on their properties . By systematically testing how UNQ511/PRO1026 affects the kinetics of different signaling pathways, researchers can determine whether it functions as a ligand, receptor, co-receptor, or signal transduction component, providing crucial insights into its biological role.
Selection of optimal expression systems for UNQ511/PRO1026 requires careful consideration of the protein's predicted properties. For mammalian expression, pcDNA3.1 vectors with sequence-optimized inserts have proven effective for uncharacterized proteins . When designing the expression construct, researchers should:
Include a signal peptide if the protein is predicted to be secreted
Consider removing transmembrane domains for improved solubility using constructs like pcDNA3.1-FΔTM
Add appropriate purification tags (His, FLAG, or Myc) at either N- or C-terminus based on predicted domain structures
Optimize codons for the expression system using algorithms that:
For PCR amplification of optimized constructs, design primers with appropriate restriction sites (e.g., KpnI/NotI) for directional cloning . After expression, purify using affinity chromatography followed by size exclusion chromatography to ensure homogeneity. Formulate the purified protein in PBS and lyophilize from a 0.2 μm filtered solution for maximum stability .
Reproducibility in studies of uncharacterized proteins presents significant challenges that must be systematically addressed. First, establish detailed standard operating procedures (SOPs) for all experimental steps, including expression, purification, and functional assays. For mass spectrometry-based identification, standardize parameters such as mass tolerances (10 ppm for precursors and 0.5 Da for fragments) and modification settings (carbamidomethylation of cysteine as fixed, oxidation of methionine as variable) .
To ensure consistent protein identification:
Require at least two detected peptides with at least one unique peptide
Set false discovery rate (FDR) cut-off to 1% for both peptides and proteins
Document all protein identifications including canonical forms, splice forms, and master forms
Validate key findings using orthogonal techniques (e.g., immunoblotting, targeted MS)
When analyzing interaction data, account for experimental variables that may affect outcomes. In BioPlex studies, among the baits represented initially by canonical sequence, for 37.6% of genes there was no information about protein sequences resulting from alternative splicing . This highlights the importance of comprehensive isoform analysis for reproducible characterization of uncharacterized proteins like UNQ511/PRO1026.
Determining subcellular localization of uncharacterized proteins requires a multi-method approach for conclusive results. For UNQ511/PRO1026, researchers should employ:
Fluorescence microscopy techniques:
Construct GFP/mCherry fusion proteins at both N- and C-termini
Perform co-localization studies with established organelle markers
Utilize super-resolution microscopy for precise spatial resolution
Biochemical fractionation:
Conduct differential centrifugation to isolate cellular compartments
Perform Western blotting of fractions using antibodies against the recombinant protein
Include compartment-specific markers to validate fractionation quality
Proximity labeling approaches:
Generate BioID or APEX2 fusion constructs
Identify proximal proteins through mass spectrometry
Compare proximity profiles with known compartment-specific proteins
Computational prediction validation:
Test predictions from algorithms like DeepLoc, TargetP, and PSORT
Experimentally validate targeting signals through deletion/mutation analysis
Assess potential dual localization patterns under different cellular conditions
Integrating these approaches provides a comprehensive view of UNQ511/PRO1026 localization, which is essential for functional hypothesis generation. Notably, discrepancies between methods may reveal dynamic localization patterns or splice variant-specific localizations, as observed in studies of other uncharacterized proteins .
Predicting functions of uncharacterized proteins like UNQ511/PRO1026 requires comprehensive bioinformatic analysis integrating multiple computational approaches. Begin with sequence-based analyses including:
Homology detection using PSI-BLAST, HHpred, and HMMER to identify distant relatives
Domain prediction using InterPro, SMART, and Pfam databases
Secondary structure prediction using PSIPRED and JPred
Disorder prediction using IUPred and PONDR
Post-translational modification site prediction using NetPhos, NetOGlyc, and NetNGlyc
Complement sequence analysis with structure-based approaches:
Ab initio structure prediction using AlphaFold2 or RoseTTAFold
Binding site prediction using CASTp and COACH
Molecular docking simulations to test interaction hypotheses
Finally, incorporate network-based approaches that have proven successful for uPE1 proteins:
Interolog mapping - transferring interactions from homologous proteins
Co-expression analysis using RNA-seq databases
Phylogenetic profiling to identify functionally related proteins
Through integration of these approaches, researchers successfully predicted Gene Ontology categories for 387 uPE1 genes in previous studies . Apply similar methodology to UNQ511/PRO1026 to generate testable functional hypotheses.
Mass spectrometry (MS) analysis for uncharacterized proteins requires specialized approaches to ensure confident identification and characterization. For UNQ511/PRO1026, follow these methodological guidelines:
Sample preparation optimization:
Employ multiple proteolytic enzymes (trypsin, chymotrypsin, GluC) to increase coverage
Implement enrichment strategies for post-translational modifications
Use FASP (Filter-Aided Sample Preparation) protocol for membrane-associated proteins
Instrument parameters:
Identification criteria:
Data analysis workflow:
Compare identified peptides against canonical, splice, and master forms
Analyze post-translational modifications with site localization scoring
Quantify relative abundance using label-free or labeled approaches
This rigorous MS methodology has successfully identified 550 uPE1 proteins in previous studies , providing a validated framework for UNQ511/PRO1026 characterization.
Protein-protein interaction (PPI) data provides crucial insights for functional annotation of uncharacterized proteins. When interpreting PPI data for UNQ511/PRO1026, researchers should implement a systematic analytical framework:
Network construction:
Build comprehensive interaction networks including direct and indirect interactions
Weight interactions based on detection confidence and reproducibility
Compare interactions across different experimental conditions and cell types
Functional inference approaches:
Apply majority rule - assign functions shared by multiple interacting partners
Implement random walk algorithms to propagate functional annotations
Calculate semantic similarity between Gene Ontology terms of interactors
Statistical validation:
Calculate enrichment scores for biological processes, cellular components, and molecular functions
Implement permutation tests to assess significance of functional predictions
Apply machine learning approaches to integrate multiple evidence types
Visualization and interpretation:
Generate network visualizations highlighting functional clusters
Calculate network metrics (degree, betweenness, clustering coefficient)
Identify potential protein complexes using algorithms like MCODE or ClusterONE
This approach has proven effective in previous studies where PPI networks with 9,967 vertices connected by 287,474 interactions successfully predicted functions for hundreds of uncharacterized proteins . For UNQ511/PRO1026, focus on interactions that are reproducible across multiple experiments and consistent with subcellular localization data.
Validating predicted interactions of uncharacterized proteins requires robust in vitro binding assays with appropriate controls. For UNQ511/PRO1026, employ a multi-tiered validation approach:
Primary binding assays:
Surface Plasmon Resonance (SPR) to determine binding kinetics (ka, kd) and affinity (KD)
Bio-Layer Interferometry (BLI) for real-time interaction analysis
Isothermal Titration Calorimetry (ITC) to measure thermodynamic parameters
Protein-based validation methods:
Pull-down assays using recombinant proteins with different tags
Co-immunoprecipitation from cell lysates expressing both interacting partners
Proximity Ligation Assay (PLA) to detect interactions in situ
Interaction specificity controls:
Test binding against structurally similar but functionally distinct proteins
Create domain deletion mutants to identify specific binding regions
Employ competitive binding assays with predicted binding partners
Functional validation:
When designing these assays, carefully consider buffer conditions, protein concentration ranges, and potential non-specific binding. For recombinant proteins, carrier-free formulations are recommended for binding assays to avoid interference from carrier proteins .
Cell-based functional validation provides critical evidence for the biological roles of uncharacterized proteins. For UNQ511/PRO1026, design comprehensive cellular assays based on bioinformatic predictions and PPI data:
Gene modulation approaches:
CRISPR-Cas9 knockout/knockdown to assess loss-of-function phenotypes
Overexpression studies using wild-type and mutant constructs
Inducible expression systems to study temporal effects
Pathway-specific reporter assays:
Luciferase reporters for transcriptional effects
FRET/BRET biosensors for real-time signaling dynamics
Calcium mobilization assays for signaling responses
Phenotypic assays based on predicted functions:
Proliferation and viability measurements
Morphological analyses using high-content imaging
Migration/invasion assays if relevant to predicted function
Specialized assays targeted to specific biological processes
Rescue experiments:
When designing these experiments, include appropriate positive and negative controls, and ensure statistical power through adequate replication. These approaches have successfully validated functions for previously uncharacterized proteins and can be adapted for UNQ511/PRO1026 based on specific functional predictions .
Research on uncharacterized proteins like UNQ511/PRO1026 presents numerous challenges that require careful methodological considerations. Common pitfalls include:
Expression and purification challenges:
Protein misfolding due to incorrect expression systems
Inadequate solubility due to hydrophobic regions
Tag interference with native protein functions
Batch-to-batch variability affecting reproducibility
Functional characterization errors:
Experimental design limitations:
Data interpretation challenges:
Conflation of correlation with causation
Confirmation bias when analyzing results
Overlooking contradictory evidence
Insufficient integration of multiple data types
To address these pitfalls, researchers should employ rigorous controls, utilize multiple orthogonal approaches, consider alternative splicing, and implement robust statistical analysis when characterizing UNQ511/PRO1026 or other uncharacterized proteins.
Accelerating functional annotation of uncharacterized proteins requires integrated strategies leveraging advanced technologies and collaborative approaches. For UNQ511/PRO1026 and similar proteins, researchers should:
Implement high-throughput screening platforms:
CRISPR-based functional genomics screens
Phenotypic screens using cell-based assays
Automated protein interaction screening
Utilize integrative bioinformatics:
Machine learning approaches combining multiple data types
Network-based function prediction algorithms
Structural modeling integrated with interaction data
Evolutionary analysis to identify conserved functional elements
Develop targeted biotechnology tools:
Establish collaborative frameworks: