Recombinant Human Uncharacterized protein C2orf66 (C2orf66)

Shipped with Ice Packs
In Stock

Description

Introduction to Recombinant Human Uncharacterized Protein C2orf66 (C2orf66)

Recombinant Human Uncharacterized Protein C2orf66, commonly referred to as C2orf66, is a protein encoded by the C2orf66 gene located on chromosome 2 in humans. Despite its designation as "uncharacterized," this protein is part of ongoing research efforts aimed at understanding its potential functions and implications in human biology and disease.

Gene and Protein Overview

  • Gene Location: The C2orf66 gene is situated on chromosome 2, which is one of the largest human chromosomes and contains numerous genes involved in various biological processes .

  • Protein Function: Currently, the specific biological functions of the C2orf66 protein remain largely uncharacterized. This means that while it is known to be a protein-coding gene, its precise role in cellular processes or disease mechanisms is not well understood .

  • Recombinant Protein: Recombinant proteins are produced through genetic engineering techniques where the gene encoding the protein is inserted into an expression vector and then expressed in a host organism (e.g., bacteria, yeast, or mammalian cells). This allows for large-scale production of the protein for research or therapeutic purposes .

Example Table: Hypothetical Research Directions for C2orf66

Research AreaMethodologyPotential Outcomes
BioinformaticsSequence alignment, structural predictionInsights into potential protein functions or interactions
Cellular StudiesImmunofluorescence, Western blottingUnderstanding protein localization and expression patterns
Functional AssaysCell culture experiments, knockout modelsDetermining the protein's role in cellular processes or disease

References

  1. NCBI Gene Database: For information on the C2orf66 gene and its genomic context.

  2. UniProt: Provides details on the protein sequence and any known functional data.

  3. GeneCards: Offers comprehensive information on the gene, including potential pathways and disorders.

  4. MyBioSource: Supplies recombinant C2orf66 protein for research purposes.

Product Specs

Form
Lyophilized powder.
Note: While we prioritize shipping the format currently in stock, please specify your format preference during order placement for customized preparation.
Lead Time
Delivery times vary depending on the purchase method and location. Please contact your local distributor for precise delivery estimates.
Note: Standard shipping includes blue ice packs. Dry ice shipping requires prior arrangement 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%, 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. Aliquot for multiple uses to prevent repeated freeze-thaw cycles.
Tag Info
The tag type is determined during the manufacturing process.
The specific tag type is determined during production. If a specific tag is required, please inform us, and we will prioritize its development.
Synonyms
C2orf66; CB066_HUMAN; Uncharacterized protein C2orf66
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
39-117
Protein Length
Full Length of Mature Protein
Purity
>85% (SDS-PAGE)
Species
Homo sapiens (Human)
Target Names
C2orf66
Target Protein Sequence
AT VRNEDKWKPL NNPRNRDLFF RRLQAYFKGR GLDLGTFPNP FPTNENPRPL SFQSELTASA SADYEEQKNS FHNYLKG
Uniprot No.

Target Background

Database Links

HGNC: 33809

KEGG: hsa:401027

STRING: 9606.ENSP00000339384

UniGene: Hs.245666

Subcellular Location
Secreted.

Q&A

What is the current annotation status of C2orf66?

C2orf66 remains classified as an uncharacterized protein encoded by a gene located on chromosome 2 in humans. Like many proteins studied in immunological contexts, its functional characterization requires systematic experimental approaches combining genetic, biochemical, and cell biological methods. When approaching uncharacterized proteins, researchers typically begin with sequence analysis tools to identify conserved domains and potential functional motifs, similar to approaches used for characterizing novel immune signaling proteins. Protein prediction algorithms can suggest potential structural elements, but these must be validated experimentally through techniques like those used to characterize TLR-interacting proteins.

What expression systems are most effective for producing recombinant C2orf66?

When producing recombinant uncharacterized proteins like C2orf66, selection of an appropriate expression system is critical. For initial characterization studies, bacterial expression systems (E. coli) offer high yield but may not provide proper folding or post-translational modifications. For functional studies, mammalian expression systems are preferred despite lower yields.

The methodology for mammalian cell expression typically involves:

  • Cloning the C2orf66 gene into a mammalian expression vector with appropriate promoter

  • Transfecting HEK293T or CHO cells using optimized transfection protocols

  • Selecting stable transfectants using antibiotic selection

  • Verifying expression using Western blotting

This approach parallels methods used for expressing recombinant human cytokines in research settings, where proper folding and post-translational modifications are critical for functional studies.

What purification strategies yield the highest purity recombinant C2orf66?

Purification of recombinant C2orf66 typically employs affinity chromatography approaches, with methodology varying based on the expression system and fusion tags employed:

Purification StepMethodologyExpected OutcomeQuality Control
Affinity ChromatographyNi-NTA for His-tagged C2orf66>80% puritySDS-PAGE
Size ExclusionSuperdex 200 column>90% purity, removal of aggregatesDynamic light scattering
Ion ExchangeResource Q column>95% purityProtein staining
Endotoxin RemovalPolymyxin B columns<0.1 EU/μg proteinLAL assay

For functional studies, ensuring removal of endotoxin contamination is particularly important as it can activate TLR4 signaling pathways and confound experimental results, similar to concerns when studying pathogen recognition receptor pathways.

How can I validate the identity and integrity of purified C2orf66?

Validation of recombinant C2orf66 requires multiple analytical approaches:

  • SDS-PAGE and Western blotting with anti-tag antibodies for initial confirmation

  • Mass spectrometry for definitive protein identification and assessment of modifications

  • Circular dichroism to confirm proper secondary structure formation

  • Size exclusion chromatography to assess oligomeric state

  • Thermal shift assays to determine protein stability

These approaches provide complementary evidence of protein identity and structural integrity before proceeding to functional studies.

What approaches can determine potential binding partners of C2orf66?

Identifying interaction partners represents a critical step in characterizing uncharacterized proteins like C2orf66. Several complementary approaches should be employed:

ApproachMethodologyAdvantagesLimitations
Yeast Two-HybridScreen against human cDNA librariesUnbiased discoveryHigh false-positive rate
Co-ImmunoprecipitationPull-down with anti-tag antibodiesDetects native complexesRequires high-quality antibodies
Proximity LabelingBioID or APEX2 fusion constructsMaps protein neighborhoodsRequires genetic manipulation
Crosslinking MSChemical crosslinking with MS analysisCaptures transient interactionsComplex data analysis
Surface Plasmon ResonanceImmobilized C2orf66 with candidate ligandsQuantitative binding kineticsRequires candidate selection

These methodologies parallel approaches used to identify novel binding partners in immune signaling pathways. For example, similar techniques revealed that bacterial tRNA can interact with TLR8 to trigger innate immune responses, demonstrating how binding partner identification can reveal unexpected functional roles.

How can I determine if C2orf66 plays a role in cellular signaling pathways?

Investigating potential signaling roles requires systematic perturbation studies:

  • Generate cell lines overexpressing C2orf66 and measure activation of common signaling pathways (NF-κB, MAPK, JAK-STAT) using reporter assays

  • Perform C2orf66 knockdown or knockout studies using siRNA or CRISPR/Cas9

  • Conduct phosphoproteomic analysis comparing wild-type and C2orf66-deficient cells

  • Examine C2orf66 expression changes during cellular activation with various stimuli

This methodological approach parallels studies of novel immune signaling components, where researchers examine how protein perturbation affects downstream signaling cascades. For instance, studies of TLR8 signaling revealed its unique ability to induce secretion of IL-12p70 in the absence of signals from other receptors, demonstrating how targeted signaling studies can reveal specialized functions.

What computational approaches can predict functional domains in C2orf66?

Computational analysis provides crucial guidance for experimental studies of uncharacterized proteins:

Computational ApproachTool ExamplesInformation Provided
Sequence HomologyBLAST, HHpredRelated proteins with known functions
Domain PredictionInterProScan, SMARTConserved functional domains
Structural ModelingAlphaFold2, I-TASSERPredicted 3D structure
Intrinsic DisorderPONDR, IUPredRegions lacking stable structure
Post-translational ModificationNetPhos, GPSPotential modification sites
Subcellular LocalizationDeepLoc, PSORTPredicted cellular compartment

These computational predictions should guide experimental design but require validation. This parallels approaches used in characterizing novel immune signaling components, where computational analysis often provides initial hypotheses about protein function that are subsequently tested experimentally.

What cell types and tissues should be prioritized for studying C2orf66 function?

Selection of relevant biological systems is critical for functional characterization:

  • Begin with expression analysis across tissue and cell types using publicly available RNA-seq databases

  • Prioritize cell types with highest expression

  • Consider disease contexts where C2orf66 expression is altered

  • Include immune cells (monocytes, macrophages, dendritic cells) given the importance of protein discovery in immune contexts

  • Develop reporter cell lines from relevant cell types

This systematic approach to biological system selection parallels successful strategies used in characterizing novel components of immune signaling pathways, where understanding cell type-specific expression patterns provides crucial context for functional studies.

How can CRISPR/Cas9 technology be optimized for studying C2orf66 function?

CRISPR/Cas9 approaches offer powerful tools for functional characterization:

  • Design multiple sgRNAs targeting different exons of C2orf66 to ensure complete knockout

  • Create tagged versions of C2orf66 at endogenous loci using HDR-mediated knock-in

  • Generate conditional knockout models to study tissue-specific functions

  • Perform CRISPR screens to identify genetic interactions

  • Use CRISPRi/CRISPRa to modulate expression without complete deletion

The guide RNA design process should prioritize specificity while minimizing off-target effects:

ConsiderationMethodologyValidation Approach
Target SelectionTarget conserved exons, avoid alternative splice sitesSequence verification
Off-target PredictionUse algorithms (CRISPOR, CHOPCHOP)Whole genome sequencing
Delivery MethodLentiviral vs. nucleofection based on cell typeDelivery efficiency measurement
Clonal SelectionSingle-cell sorting and expansionGenotyping PCR and sequencing
Phenotype ValidationRescue experiments with wildtype C2orf66Functional assays

What approaches can resolve contradictory findings about C2orf66 function?

When facing conflicting experimental results, systematic troubleshooting is essential:

  • Carefully examine differences in experimental systems (cell types, stimulation conditions)

  • Verify reagent quality and specificity, particularly antibodies

  • Implement orthogonal methodologies to validate findings

  • Consider genetic background effects in different cell lines

  • Evaluate protein interaction networks in different contexts

These approaches parallel strategies used in resolving contradictory findings in immune signaling research. For instance, apparently conflicting reports about TLR8 polymorphisms and tuberculosis susceptibility were resolved through meta-analysis and population-specific studies, demonstrating how methodological rigor can clarify seemingly contradictory results.

What controls are essential when studying an uncharacterized protein like C2orf66?

Rigorous control implementation is critical for reliable characterization:

  • Use multiple cell lines to avoid cell type-specific artifacts

  • Include empty vector/untransfected controls in overexpression studies

  • Employ scrambled/non-targeting controls in knockdown experiments

  • Validate knockout phenotypes with rescue experiments

  • Include positive controls for pathway activation in signaling studies

  • Implement biological replicates across independent experiments

These control strategies parallel approaches used in immune signaling research, where careful control implementation is essential for distinguishing specific effects from experimental artifacts.

How can post-translational modifications of C2orf66 be comprehensively mapped?

Post-translational modification (PTM) analysis requires integrated approaches:

PTM TypeDetection MethodologyFunctional Validation
PhosphorylationPhospho-specific antibodies, LC-MS/MS with phosphopeptide enrichmentPhosphomimetic and phosphodeficient mutants
UbiquitinationUbiquitin pull-down, K-ε-GG antibody enrichmentLysine-to-arginine mutants
GlycosylationGlycosidase treatment, lectin binding, glycoproteomic MSN/O-glycosylation site mutants
SUMOylationSUMO-IP, MS analysisConsensus site mutations
AcetylationAnti-acetyl-lysine antibodies, MSLysine-to-arginine mutants

Understanding PTMs can provide critical insights into protein regulation and function, as demonstrated in studies of immune signaling proteins where phosphorylation events often serve as molecular switches that control pathway activation.

What are the best strategies for generating specific antibodies against C2orf66?

Development of specific antibodies against uncharacterized proteins requires careful design:

  • Perform epitope prediction to identify antigenic regions unique to C2orf66

  • Generate multiple antibodies against different regions (N-terminal, C-terminal, internal domains)

  • Use both peptide and recombinant protein immunization strategies

  • Rigorously validate antibody specificity using knockout cells and overexpression controls

  • Characterize each antibody for specific applications (Western blot, IP, flow cytometry)

These approaches parallel successful strategies used in developing antibodies against novel immune signaling components, where antibody specificity is critical for accurately interpreting experimental results.

How might C2orf66 function relate to human disease pathways?

Connecting uncharacterized proteins to disease contexts requires integrative approaches:

  • Analyze expression patterns in disease-relevant tissues using public databases

  • Examine genetic variants in C2orf66 associated with disease phenotypes

  • Investigate protein interactions with known disease-associated proteins

  • Perform pathway analysis to position C2orf66 within known disease networks

  • Consider evolutionary conservation as an indicator of functional importance

This systematic approach parallels strategies used to connect novel immune signaling components to disease processes. For example, polymorphisms in TLR8 were associated with altered progression of HIV and tuberculosis through genome-wide association studies, demonstrating how genetic approaches can reveal unexpected disease connections.

What model systems are most appropriate for studying C2orf66 in a disease context?

Selection of appropriate model systems is critical for translational studies:

Model SystemAdvantagesLimitationsBest Applications
Cell LinesGenetic manipulation, high throughputLimited physiological relevanceMechanistic studies, initial screening
Primary Human CellsPhysiological relevanceDonor variability, limited lifespanValidation of mechanisms in human context
Mouse ModelsIn vivo systems, genetic manipulationSpecies differencesPhysiological studies, disease models
Patient SamplesDirect disease relevanceLimited availability, experimental constraintsCorrelation studies, biomarker validation
Organoids3D architecture, tissue-specific functionsTechnical challenges, limited throughputComplex cellular interactions

The selection of appropriate model systems should be guided by the specific research question and the translational goals of the study.

How can I overcome common challenges in C2orf66 expression and purification?

Troubleshooting strategies for difficult-to-express proteins include:

  • Optimize codon usage for the expression system

  • Test multiple fusion tags and their placement (N-terminal vs. C-terminal)

  • Evaluate different cell lysis and extraction buffers

  • Consider expressing individual domains if full-length protein is unstable

  • Implement co-expression with potential binding partners

  • Test expression at lower temperatures to improve folding

These approaches parallel strategies used to overcome expression challenges with immune signaling proteins, where protein structure and stability often present technical hurdles.

What strategies can resolve inconsistent results in C2orf66 functional assays?

When facing experimental inconsistencies:

  • Standardize experimental protocols across all studies

  • Validate key reagents, particularly antibodies and recombinant proteins

  • Implement positive and negative controls in each experiment

  • Consider cell density, passage number, and culture conditions as variables

  • Evaluate timing of measurements for dynamic processes

  • Employ multiple methodologies to measure the same outcome

These troubleshooting approaches reflect best practices in immune signaling research, where experimental standardization is essential for generating reproducible results.

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.