Cyclophilin-E, a member of the peptidyl-prolyl cis-trans isomerase (PPIase) family, catalyzes the cis-trans isomerization of proline imidic peptide bonds in oligopeptides, accelerating protein folding. This protein possesses a highly conserved cyclophilin domain and an RNA-binding domain, contributing to its PPIase activity, protein folding capabilities, and RNA-binding activity. Notably, Cyclophilin-E features two RNA binding domains at its N-terminal region and a PPIase domain at its C-terminal region.
The formulation contains 1mg/ml of Cyclophilin-E antibody in a buffer consisting of PBS at pH 7.4, 10% Glycerol, and 0.02% Sodium Azide.
The Cyclophilin-E antibody has undergone rigorous testing through ELISA and Western blot analysis, ensuring its specificity and reactivity. However, due to variations in applications, it is recommended to titrate the reagent for each specific experiment to achieve optimal results.
Peptidyl-prolyl cis-trans isomerase E, PPIase E, Rotamase E, Cyclophilin-33, PPIE, peptidylprolyl isomerase E, CYP33, Cyclophilin E, CYP-33, MGC3736, MGC111222.
Cyclophilin-E antibody was purified from mouse ascitic fluids by protein-A affinity chromatography.
PAT17E8AT.
Anti-human Cyclophilin-E mAb, is derived from hybridization of mouse F0 myeloma cells with spleen cells from BALB/c mice immunized with recombinant human Cyclophilin-E amino acids 1-301 purified from E. coli.
Mouse IgG1 heavy chain and κ light chain.
Cyclophilin-E (also known as CyPE, Cyclophilin 33, CyP33, PPIase E, or Rotamase E) is a crucial nuclear RNA-binding protein involved in significant cellular processes. It features a unique structure with an N-terminal RNA binding domain and a C-terminal cyclophilin domain, allowing specific interactions with mRNA. This structural configuration enhances its peptidyl-prolyl isomerase activity, facilitating proper protein folding and gene expression regulation .
Cyclophilin-E is particularly important in research because it binds to the third PHD zinc finger domain of Mixed Lineage Leukemia (MLL) protein, influencing transcriptional regulation of target genes including HoxC8 and HoxC9. Its overexpression can inhibit transcription of these genes by promoting histone deacetylase 1 binding to MLL repression domain . Additionally, it functions in pre-mRNA splicing as a component of the spliceosome and demonstrates preference for single-stranded RNA molecules with poly-A and poly-U stretches .
Several types of Cyclophilin-E antibodies are available, each with specific characteristics suitable for different research applications:
The choice of antibody depends on the specific research application, species of interest, and detection method required.
Cyclophilin-E differs significantly from other cyclophilins such as Cyclophilin A in structure, cellular localization, and function:
Structure and Domains: Cyclophilin-E uniquely contains both an RNA-binding domain (RRM) at its N-terminus and a cyclophilin domain at its C-terminus . In contrast, Cyclophilin A is smaller (~18 kDa) and lacks the RNA-binding domain .
Cellular Localization: Cyclophilin-E is predominantly nuclear and participates in nuclear processes like pre-mRNA splicing . Cyclophilin A is ubiquitously distributed intracellularly and can be secreted by cells in response to inflammatory stimuli .
Functional Roles: While both possess peptidyl-prolyl isomerase activity, Cyclophilin-E specifically regulates transcription through interactions with MLL protein and demonstrates antiviral properties against influenza virus by interacting with viral nucleoprotein . Cyclophilin A interacts with HIV proteins and is necessary for infectious HIV virion formation .
Research Applications: Antibodies against these different cyclophilins target distinct biological processes and pathways, making them valuable for specific research questions related to either nuclear gene regulation (Cyclophilin-E) or cytoplasmic and secretory pathways (Cyclophilin A).
Optimizing Western blotting with Cyclophilin-E antibodies requires careful attention to several parameters:
Sample Preparation and Loading:
Use fresh lysates when possible, with complete protease inhibitor cocktails
For Cyclophilin-E detection, a concentration range of 0.04-0.4 μg/ml is recommended for polyclonal antibodies
Include positive controls such as lysates from cells known to express Cyclophilin-E (HEK293T cells with PPIE overexpression work well)
Blocking and Antibody Incubation:
For monoclonal antibodies like clone 9E18, use standard blocking solutions (5% non-fat milk or BSA)
Primary antibody incubation is typically performed overnight at 4°C
For the rabbit polyclonal antibody (NBP1-85365), optimal dilution ranges from 1:50 to 1:200 for immunohistochemistry and 0.04-0.4 μg/ml for Western blotting
Validation Controls:
Include a vector-only transfected control alongside PPIE overexpression lysate to confirm specificity
The Cyclophilin-E protein should be detected at approximately 33 kDa
For tagged proteins, adjust the expected molecular weight accordingly (e.g., C-terminal myc-DDK tag adds 3.1 kDa)
Western blot analysis has successfully demonstrated specificity of anti-Cyclophilin-E antibodies by comparing control (vector-only transfected HEK293T lysate) with PPIE overexpression lysate , confirming the antibody's ability to specifically detect the target protein.
Successful immunohistochemical detection of Cyclophilin-E requires careful optimization:
Tissue Preparation and Antigen Retrieval:
For paraffin-embedded sections, heat-induced epitope retrieval (HIER) at pH 6 is specifically recommended
Complete deparaffinization and rehydration are essential for consistent results
Antibody Selection and Dilution:
For polyclonal antibodies like NBP1-85365, optimal dilution ranges from 1:50 to 1:200
Incubation times may require optimization (typically overnight at 4°C or 1-2 hours at room temperature)
Signal Detection and Interpretation:
Cyclophilin-E typically shows strong nuclear positivity, as demonstrated in human cerebellum samples
Include positive tissue controls with known Cyclophilin-E expression
Consider dual staining with markers of cellular compartments to confirm localization
When properly optimized, immunohistochemistry with anti-Cyclophilin-E antibody reveals strong nuclear localization pattern, consistent with its known role in nuclear processes such as pre-mRNA splicing and transcriptional regulation .
Rigorous controls are essential for reliable interpretation of experiments using Cyclophilin-E antibodies:
Positive Controls:
Lysates or tissues with confirmed Cyclophilin-E expression
Recombinant Cyclophilin-E protein can serve as a positive control in Western blots
HEK293T cells with PPIE overexpression provide an excellent positive control
Negative Controls:
Primary antibody omission to assess non-specific binding of secondary antibodies
Isotype-matched control antibodies (especially important for monoclonal antibodies like 9E18)
For siRNA experiments, non-targeting siRNA controls should be used as demonstrated in influenza virus studies
Specificity Controls:
Comparison between wild-type and Cyclophilin-E knockout/knockdown samples
Pre-adsorption with recombinant Cyclophilin-E protein
Testing on multiple expression systems and cell lines to ensure consistent results
Functional Validation:
For studies investigating Cyclophilin-E's PPIase activity, include wild-type protein and catalytically inactive mutants
When studying protein-protein interactions (like with MLL), include domain deletion mutants to confirm specificity of binding regions
In published research, specificity of anti-Cyclophilin-E antibodies has been validated using overexpression systems, comparing vector-only controls with Cyclophilin-E-expressing cells, and through domain deletion studies that confirm functional specificity .
Cyclophilin-E has emerged as an important host factor in viral infections, particularly influenza virus. Antibodies against Cyclophilin-E can be strategically employed to investigate these interactions:
Co-Immunoprecipitation Studies:
Anti-Cyclophilin-E antibodies have been used to demonstrate direct interaction between Cyclophilin-E and viral proteins
In influenza research, cell lysates immunoblotted with anti-Myc or anti-FLAG antibodies confirmed expression of proteins of interest in 293T cells
These studies revealed that Cyclophilin-E functions as a negative regulator to influenza virus by impairing the formation of viral ribonucleoprotein (vRNP)
Functional Validation Through Knockdown/Overexpression:
Knockdown of endogenous Cyclophilin-E has been shown to favor influenza virus replication
Conversely, overexpression of Cyclophilin-E decreased viral nucleoprotein levels and reduced virus titers approximately twofold compared to control cells
Importantly, this antiviral effect was dependent on Cyclophilin-E binding to viral nucleoprotein, as demonstrated using the Cyclophilin-E Δ137–186 deletion mutant
Methodological Approach:
Perform co-immunoprecipitation using anti-Cyclophilin-E antibodies to pull down viral protein complexes
Use Western blotting with anti-Cyclophilin-E antibodies to monitor expression levels in knockdown or overexpression experiments
Complement with immunofluorescence studies to visualize subcellular localization during infection
Utilize domain mutants to map specific interaction regions between Cyclophilin-E and viral proteins
These approaches have established Cyclophilin-E as a host restriction factor that targets viral nucleoprotein functions, providing a potential target for antiviral strategies .
Studying Cyclophilin-E's interaction with the MLL (Mixed Lineage Leukemia) protein complex requires sophisticated methodological approaches:
Co-Immunoprecipitation and Pulldown Assays:
Anti-Cyclophilin-E antibodies can be used to precipitate native protein complexes containing MLL
These experiments have revealed that Cyclophilin-E binds to the third PHD zinc finger domain of MLL protein
This interaction influences transcriptional regulation of target genes including HoxC8 and HoxC9
Functional Analysis Through Gene Expression Studies:
Cyclophilin-E overexpression inhibits transcription of MLL target genes by promoting histone deacetylase 1 binding to MLL repression domain
Researchers can use anti-Cyclophilin-E antibodies in chromatin immunoprecipitation (ChIP) assays to detect recruitment to specific genomic loci
Domain Mapping and Mutational Analysis:
The peptidyl-prolyl isomerase activity of Cyclophilin-E is required for inhibition of KMT2A (MLL) activity
Strategic use of domain-specific antibodies or epitope-tagged constructs can help map the specific regions involved in these interactions
Methodological Workflow:
Perform co-IP experiments using anti-Cyclophilin-E antibodies to confirm interaction with MLL complex components
Use ChIP with anti-Cyclophilin-E to identify genomic binding sites
Combine with gene expression analysis after Cyclophilin-E knockdown or overexpression
Validate with domain mutants and functional readouts of MLL activity
These approaches have established Cyclophilin-E as an important regulator of MLL function, with significant implications for understanding gene regulation in developmental processes and diseases .
Cyclophilin-E's involvement in pre-mRNA splicing as a spliceosome component makes its antibodies valuable tools for investigating this fundamental cellular process:
Spliceosome Complex Analysis:
Anti-Cyclophilin-E antibodies can immunoprecipitate spliceosome complexes for proteomic analysis
This approach has confirmed Cyclophilin-E as a component of the spliceosome involved in pre-mRNA splicing
RNA-Binding Studies:
Cyclophilin-E combines RNA-binding and PPIase activities, with preference for single-stranded RNA molecules containing poly-A and poly-U stretches
RNA immunoprecipitation (RIP) with anti-Cyclophilin-E antibodies can identify bound RNA species
These experiments suggest Cyclophilin-E binds to the poly(A)-region in the 3'-UTR of mRNA molecules
Functional Splicing Assays:
Depletion or inhibition of Cyclophilin-E can be monitored using Western blotting with anti-Cyclophilin-E antibodies
Effects on splicing can be assessed using minigene reporters or RNA-seq to detect alterations in splicing patterns
Methodological Strategy:
Utilize anti-Cyclophilin-E antibodies for immunofluorescence to co-localize with other splicing factors
Perform RIP-seq to identify RNA targets bound by Cyclophilin-E
Use knockdown/overexpression approaches with splicing-sensitive reporters
Complement with in vitro splicing assays using immunodepleted nuclear extracts
These approaches provide insights into how Cyclophilin-E's dual RNA-binding and PPIase activities contribute to proper pre-mRNA processing, with implications for understanding splicing regulation in normal and disease states .
Researchers using Cyclophilin-E antibodies in Western blotting may encounter several challenges:
High Background or Non-specific Bands:
Problem: Multiple bands or high background obscuring the specific Cyclophilin-E signal
Solution: Increase blocking time/concentration, optimize antibody dilution (0.04-0.4 μg/ml recommended for polyclonal antibodies) , use more stringent washing conditions, and consider alternative blocking reagents
Weak or No Signal:
Problem: Failure to detect Cyclophilin-E despite predicted expression
Solution: Check sample preparation (fresh lysates with protease inhibitors), increase protein loading, optimize transfer conditions for proteins in the 33 kDa range, and consider enhanced chemiluminescence detection systems
Inconsistent Results Between Samples:
Problem: Variable detection across similar samples
Solution: Standardize protein quantification methods, ensure equal loading using housekeeping controls, and maintain consistent sample preparation protocols
Band Size Discrepancies:
Problem: Detected band at unexpected molecular weight
Solution: Cyclophilin-E should appear at approximately 33 kDa; post-translational modifications or splice variants may alter migration. Include positive controls like PPIE-overexpressing HEK293T cells , and consider the addition of tags when using recombinant systems (e.g., myc-DDK tag adds 3.1 kDa)
Low Reproducibility:
Problem: Results vary between experiments
Solution: Standardize protocols, use the same antibody lot when possible, and include consistent positive and negative controls in each experiment
Successfully addressing these issues enables reliable detection of Cyclophilin-E, as demonstrated in published research comparing control and PPIE-overexpressing cell lysates .
Contradictory results between different anti-Cyclophilin-E antibodies require systematic analysis:
Epitope Differences:
Different antibodies target distinct epitopes on Cyclophilin-E
The monoclonal antibody 9E18 recognizes specific epitopes that may be masked in certain experimental conditions
Polyclonal antibodies like NBP1-85365 were developed against a specific recombinant protein fragment (amino acids: EEVDDKVLHAAFIPFGDITDIQIPLDYETEKHRGFAFVEFELAEDAAAAIDNMNESELFGRTIRVNLAKPMRIKEGSSRPVWSDDDWLKKF)
Resolution Strategy:
Comparative Analysis: Test multiple antibodies side-by-side on identical samples
Validation Controls: Include Cyclophilin-E overexpression, knockdown, and knockout samples
Alternative Detection Methods: Complement antibody-based detection with mass spectrometry or RNA expression data
Domain-Specific Analysis: Consider whether results differ due to detection of specific protein domains, isoforms, or post-translational modifications
Interpretation Framework:
Consistent results across multiple antibodies provide stronger evidence
Discrepancies may reveal biologically significant information about protein conformation, interactions, or modifications
Results should be interpreted in the context of the specific experimental system and conditions
When analyzing contradictory results, researchers should consider the specific properties of each antibody, including clonality, host species, and the immunogen used for development , as these factors significantly impact detection patterns and specificity.
Non-specific staining is a common challenge in immunohistochemistry that requires systematic troubleshooting:
Optimizing Blocking Conditions:
Increase blocking time or concentration
Test alternative blocking agents (BSA, normal serum, commercial blocking reagents)
For polyclonal antibodies like NBP1-85365, thorough blocking is particularly important
Antigen Retrieval Optimization:
For paraffin sections, HIER pH 6 retrieval is specifically recommended for Cyclophilin-E detection
Adjust retrieval time and temperature based on tissue type and fixation conditions
Antibody Dilution Series:
Test a range of antibody dilutions (1:50 - 1:200 recommended for NBP1-85365)
Perform parallel staining with serial dilutions to identify optimal signal-to-noise ratio
Controls to Identify Sources of Non-specificity:
Primary antibody omission
Isotype controls
Pre-absorption with recombinant Cyclophilin-E protein
Comparison with known positive samples (human cerebellum shows strong nuclear positivity)
Signal Development Optimization:
Reduce substrate development time
Use alternative detection systems
Consider fluorescent detection for better signal discrimination
Interpretation Guidelines:
Cyclophilin-E shows predominantly nuclear localization
Cytoplasmic staining may represent non-specific binding or cross-reactivity
Pattern should be consistent with known biology (nuclear localization for transcription/splicing functions)
Proper optimization typically yields clear nuclear staining in appropriate tissues, as demonstrated with anti-Cyclophilin-E antibody in human cerebellum samples .
Cutting-edge research on Cyclophilin-E's RNA-binding functions benefits from integrating antibodies with emerging technologies:
CLIP-Seq Applications:
Cross-linking immunoprecipitation sequencing (CLIP-seq) using anti-Cyclophilin-E antibodies can map RNA binding sites with nucleotide resolution
This approach extends previous findings that Cyclophilin-E binds single-stranded RNA with preference for poly-A and poly-U stretches
CLIP-seq can identify the precise RNA motifs and structures recognized by Cyclophilin-E's RNA-binding domain
Proximity Labeling Approaches:
BioID or APEX2 fusions with Cyclophilin-E can identify proteins in proximity during RNA processing
Anti-Cyclophilin-E antibodies verify expression and localization of fusion proteins
These methods provide spatial context for Cyclophilin-E function in spliceosomes and transcriptional complexes
Single-Molecule Imaging:
Anti-Cyclophilin-E antibodies conjugated to fluorophores enable tracking of individual molecules
Combined with labeled RNA, this approach can visualize RNA-protein interactions in real-time
These techniques help understand the dynamics of Cyclophilin-E's binding to target RNAs
Cryo-EM Structural Analysis:
Antibody fragments can facilitate structure determination of Cyclophilin-E complexes
This approach may reveal conformational changes associated with RNA binding and isomerization
Structural insights complement functional data on Cyclophilin-E's dual domains
These integrated approaches extend beyond traditional applications, providing deeper insights into how Cyclophilin-E's RNA-binding and PPIase activities coordinate to regulate splicing and gene expression .
Emerging research suggests Cyclophilin-E's involvement in several disease processes, with antibodies serving as critical tools for investigation:
Viral Infections:
Cyclophilin-E functions as a negative regulator of influenza virus replication
Anti-Cyclophilin-E antibodies have demonstrated that CypE interacts with viral nucleoprotein
Overexpression studies using tagged Cyclophilin-E (detected with anti-Myc) showed decreased viral replication and approximately twofold reduction in virus titer
Importantly, this antiviral effect requires binding to nucleoprotein, as the CypE Δ137–186 deletion mutant loses this activity
Transcriptional Dysregulation:
Cyclophilin-E regulates gene expression through interaction with the MLL protein, influencing HoxC8 and HoxC9 genes
Antibodies enable chromatin immunoprecipitation to map genomic binding sites
These approaches may reveal Cyclophilin-E's involvement in developmental disorders or cancers associated with MLL dysregulation
Splicing-Related Disorders:
As a spliceosome component , Cyclophilin-E could contribute to splicing-related diseases
Anti-Cyclophilin-E antibodies can help identify aberrant spliceosome composition or localization
Immunoprecipitation followed by RNA analysis may reveal altered RNA binding in disease states
Research Applications:
Use antibodies to assess Cyclophilin-E expression levels in disease tissues
Perform co-immunoprecipitation to identify altered protein interactions in pathological states
Develop therapeutic strategies targeting Cyclophilin-E's PPIase activity or protein interactions
Employ domain-specific antibodies to distinguish functional activities in different contexts
This research direction may identify Cyclophilin-E as a therapeutic target or biomarker for viral infections and potentially other diseases involving RNA processing dysregulation .
Integrating Cyclophilin-E antibodies into multi-omics workflows requires careful methodological considerations:
Immunoprecipitation for Proteomics:
Anti-Cyclophilin-E antibodies can enrich protein complexes for mass spectrometry analysis
Consider cross-linking approaches to capture transient interactions
Compare results from multiple antibodies targeting different epitopes
Include appropriate controls (IgG, isotype controls) for accurate interaction identification
ChIP-Seq and RNA-Binding Analysis:
Optimize chromatin immunoprecipitation conditions for Cyclophilin-E's nuclear localization
Validate antibody specificity in both IP and Western blot before proceeding to sequencing
Consider sequential ChIP to identify co-occupancy with other factors
For RNA studies, RIP-seq or CLIP-seq require highly specific antibodies with minimal background
Integration with Transcriptomics:
Correlate Cyclophilin-E binding sites with transcriptome changes after manipulation
Compare RNA-seq data from wild-type and Cyclophilin-E knockdown/knockout systems
Analyze alternative splicing events that may depend on Cyclophilin-E's spliceosome function
Data Analysis Considerations:
Account for antibody efficiency/bias in computational analysis pipelines
Use appropriate normalization strategies when comparing datasets generated with different antibodies
Integrate results from complementary approaches (e.g., proteomics, genomics, transcriptomics)
Consider biological replicates to ensure reproducibility
Quality Control Metrics:
Verify antibody specificity before large-scale experiments
Include spike-in controls for quantitative analyses
Document lot-to-lot variation that may affect multi-omics data comparability
Validate key findings with orthogonal methods
These considerations enable robust multi-omics research that can comprehensively characterize Cyclophilin-E's diverse functions in pre-mRNA splicing , transcriptional regulation , and antiviral response .
Cyclophilin-E (CypE), also known as Peptidyl-prolyl cis-trans isomerase E (PPIE), is a member of the cyclophilin family of proteins. Cyclophilins are peptidyl-prolyl cis-trans isomerases (PPIases) that catalyze the cis-trans isomerization of proline imidic peptide bonds in oligopeptides, facilitating protein folding and function .
Cyclophilin-E contains a highly conserved cyclophilin (CYP) domain and an RNA-binding domain . This dual functionality allows CypE to participate in both protein folding and RNA-binding activities. The protein is involved in various cellular processes, including protein folding, signal transduction, and immune response .
Antibodies targeting Cyclophilin-E, such as mouse anti-human Cyclophilin-E antibodies, are valuable tools in scientific research. These antibodies are used in several applications, including:
Cyclophilins, including Cyclophilin-E, are implicated in various pathological states such as viral replication, inflammation, and cancer progression . Inhibition of Cyclophilin activity has been explored as a therapeutic strategy for treating diseases like hepatitis C virus (HCV) infection, human immunodeficiency virus (HIV) infection, and certain cancers . Non-immunosuppressive derivatives of cyclosporine A (CsA) have shown promise as Cyclophilin inhibitors, highlighting their potential as therapeutic agents .