Peptidyl-Prolyl Isomerase (PPIase): CypB catalyzes cis-trans isomerization of proline residues, critical for protein folding. This activity is inhibited by cyclosporin A (CsA) with an IC₅₀ of ~15 nM ( ).
Binding Affinity: Surface plasmon resonance (SPR) reveals a strong interaction with Plasmodium falciparum PfRhopH3 (K<sub>D</sub> = 1.6 × 10⁻⁷ M) ( ).
Endoplasmic Reticulum (ER): Native CypB contains an ER-directed signal sequence ( ).
Erythrocyte Surface: Immunofluorescence confirms CypB localization on human red blood cells (RBCs), where it mediates P. falciparum merozoite invasion ( ).
Receptor-Ligand Interaction: CypB binds PfRhopH3 on P. falciparum merozoites, facilitating RBC invasion. Anti-PfRhopH3 antibodies reduce CypB-merozoite binding by >50% ( ).
Multi-Protein Complex: CypB forms a complex with Basigin (CD147) and interacts with PfRh5, enhancing invasion efficiency ( ).
Cyclosporin A: Blocks merozoite invasion by disrupting CypB-PfRhopH3 binding ( ).
De Novo Peptide CDP3: A 98-residue peptide inhibits CypB-mediated invasion by 80% ( ).
CsA-CypB Complex: Inhibits calcineurin phosphatase activity, suppressing T-cell activation ( ). Structural studies show CsA binds similarly to CypB and CypA, but CypB/CsA exhibits stronger calcineurin inhibition due to surface residues (Arg90, Lys113, Ala128) ( ).
Extracellular Signaling: Secreted CypB promotes inflammation by inducing IL-6, IL-8, and adhesion molecules in immune cells ( ).
Cyclophilin B (CypB) belongs to the cyclophilin family of peptidyl-prolyl isomerases (PPIases) that accelerate protein folding by catalyzing the cis-trans isomerization of proline residues. CypB is primarily localized in the endoplasmic reticulum (ER) where it functions as a molecular chaperone assisting in protein folding and quality control . Unlike other cyclophilin family members, CypB contains an ER retention signal and can be secreted into biological fluids, where it mediates inflammatory responses through interaction with the CD147 receptor on cell surfaces . The PPIase activity of CypB is essential for its protective function against ER stress-induced cell death, as demonstrated by studies showing that isomerase activity-defective mutants (CypB/R62A) increase calcium leakage from the ER, reactive oxygen species (ROS) generation, and decrease mitochondrial membrane potential .
His-tagged human Cyclophilin B is a recombinant protein engineered with a polyhistidine tag (typically 6-10 histidine residues) at either the N- or C-terminus to facilitate purification using immobilized metal affinity chromatography (IMAC). While the His-tag provides significant research advantages, researchers should consider potential functional implications:
Structural Considerations:
The His-tag rarely interferes with the core PPIase domain structure, as it is typically attached to terminal regions
Crystal structures show that the active site of cyclophilins is formed by residues from the central β-sheet and surrounding loops, which are distant from the termini where tags are attached
Functionality Assessment:
To verify His-tagged CypB functionality, researchers should:
Perform comparative PPIase activity assays using both tagged and untagged versions
Assess binding affinity to known partners like Cyclosporin A
Evaluate chaperone activity through protein folding assays
Consider tag removal using protease cleavage if interference is detected
Recent research has revealed significant variations in Cyclophilin B serum concentrations across the menstrual cycle, suggesting hormonal regulation of this protein. A 2023 study monitoring eleven healthy women with normal BMI (21.8 kg/m²) throughout a single menstrual cycle demonstrated a specific pattern of CypB expression :
Menstrual Phase | Relative CypB Concentration | Statistical Significance |
---|---|---|
Follicular | Highest | p = 0.012 |
Periovulatory | Lowest | p = 0.012 |
Mid-luteal | Intermediate (slight increase from periovulatory) | p = 0.012 |
Methodology for investigating cycle-dependent variations:
Recruit female subjects with regular menstrual cycles
Collect blood samples at precisely defined cycle phases (confirmed by hormone measurements)
Employ ELISA or immunoblotting with anti-CypB antibodies for quantification
Analyze data using repeated measures ANOVA with post-hoc tests
Correlate CypB levels with estradiol, progesterone, FSH, and LH measurements
These findings suggest potential roles for CypB in cyclic inflammatory events affecting the female reproductive system and highlight the importance of controlling for menstrual cycle phase when measuring CypB in female subjects .
The functional distinction between CypB and other cyclophilins, particularly CypA, has significant implications for understanding their roles in disease processes. Recent research demonstrates remarkable specificity in their contributions to pathological conditions:
Non-Alcoholic Steatohepatitis (NASH) Development:
A 2024 study using knockout mouse models revealed that Ppib-/- (CypB KO) mice were protected from developing NASH features in a diet and chemical-induced model, while Ppia-/- (CypA KO) mice developed severe disease comparable to wild-type mice . This demonstrates that:
CypB plays a necessary role in NASH disease progression
CypA loss cannot compensate for CypB function in this context
The specific role of CypB in the ER secretory pathway may be significant to NASH pathogenesis
Research methodology for investigating cyclophilin isoform specificity:
Generate conditional and tissue-specific knockout models for individual cyclophilins
Apply isoform-specific inhibitors when available
Perform rescue experiments by expressing specific cyclophilin isoforms in knockout backgrounds
Use quantitative proteomics to identify isoform-specific interaction partners
Evaluate localization patterns through subcellular fractionation and immunofluorescence
This functional specificity extends to other disease contexts and should guide research design when investigating cyclophilin-associated pathologies .
Human cyclophilins share a conserved PPIase domain but exhibit distinct functional characteristics driven by specific structural differences:
Key structural distinctions of CypB:
Contains an N-terminal signal sequence directing it to the ER
Features an ER retention signal (HEEL) at its C-terminus
Possesses a core PPIase domain with specific substrate specificity
Comparative structural analysis:
The cyclophilin family shows greatest conformational divergence in the S2 pocket region, which influences substrate specificity . Structural studies reveal that cyclophilins can be categorized by:
β1-β2 loop configuration:
"Full-length" loops (PPIB/CypB, PPIC, PPID, PPIE, PPIF, PPIG, PPIH, PPIL6, NKTR, RanBP2)
"Deleted" loops (PPIL1, PPIL2, PPIL3, PPIL4, SDCCAG-10, PPWD1)
Active site residue variations:
α1-β3 loop region:
This region shows substantial structural diversity among cyclophilin family members
Influences interaction with binding partners and subcellular localization
Experimental approaches to study these structural distinctions include X-ray crystallography, NMR spectroscopy, and hydrogen-deuterium exchange mass spectrometry to map interaction surfaces and conformational dynamics .
The peptidyl-prolyl isomerase (PPIase) activity of CypB plays a critical role in protecting cells against ER stress-induced apoptosis through several mechanisms:
PPIase-dependent cytoprotection:
Research demonstrates that wild-type CypB attenuates ER stress-induced cell death, while PPIase-defective mutants (CypB/R62A) exacerbate it . The specific mechanisms include:
Protein folding acceleration:
CypB catalyzes the rate-limiting cis-trans isomerization of proline residues
This activity directly accelerates protein folding, reducing misfolded protein burden
ER calcium homeostasis maintenance:
PPIase-defective CypB increases Ca²⁺ leakage from the ER
Wild-type CypB helps maintain proper calcium storage in the ER lumen
Mitochondrial integrity preservation:
CypB PPIase activity helps maintain mitochondrial membrane potential during ER stress
This prevents activation of the intrinsic apoptotic pathway
ROS generation suppression:
Functional CypB limits reactive oxygen species production during ER stress
This protective effect requires intact PPIase activity
Chaperone network integration:
CypB interacts with other ER stress-related chaperones including BiP and Grp94
These interactions require functional PPIase domains to maintain the ER protein folding network
To investigate these mechanisms, researchers can employ experimental approaches including PPIase activity assays with purified proteins, calcium imaging in ER stress conditions, ROS detection assays, and co-immunoprecipitation studies to map interactions with other chaperones .
Purification of His-tagged human Cyclophilin B requires careful optimization to maintain structural integrity and enzymatic activity. The following protocol integrates best practices for high-yield, high-purity preparations:
Expression system selection:
E. coli BL21(DE3) for high yield of soluble protein
Mammalian expression systems (HEK293 or CHO cells) when post-translational modifications are required
Optimized purification workflow:
Immobilized Metal Affinity Chromatography (IMAC):
Use Ni-NTA or Co-TALON resins (Co²⁺ typically provides higher specificity)
Buffer composition: 50 mM Tris-HCl pH 8.0, 300 mM NaCl, 10% glycerol
Include 5-10 mM imidazole in binding buffer to reduce non-specific binding
Elute with 250-300 mM imidazole gradient
Size Exclusion Chromatography:
Further purify using Superdex 75 column
Buffer: 20 mM HEPES pH 7.5, 150 mM NaCl
Monitor for monomeric state (~21 kDa for His-tagged CypB)
Quality control assessments:
SDS-PAGE (>95% purity)
Western blot with anti-His and anti-CypB antibodies
Circular dichroism to verify secondary structure
PPIase activity assay using standard peptide substrates
Considerations for specific applications:
For crystallography: Include final polishing step with ion exchange chromatography
For functional assays: Verify activity with PPIase assay using tetrapeptide substrate
For binding studies: Consider tag removal using TEV or PreScission protease
Proper storage in 20 mM HEPES pH 7.5, 150 mM NaCl, 10% glycerol at -80°C maintains activity for >6 months. Avoid repeated freeze-thaw cycles.
Studying CypB-mediated cyclosporin A (CsA) internalization requires specialized techniques to track both the protein and drug. Based on established research methodologies , the following approach is recommended:
Preparation of CsA-CypB complex:
Incubate purified His-tagged CypB with CsA at 1:1 molar ratio
Confirm complex formation using isothermal titration calorimetry or fluorescence polarization
Binding and internalization assay:
Isolate peripheral blood T-lymphocytes using Ficoll-Hypaque gradient and negative selection
Label CypB with fluorescent tag (e.g., Alexa Fluor 488) or radiolabel (³H or ¹²⁵I)
Label CsA with distinct fluorophore (e.g., Alexa Fluor 647) or tritium (³H-CsA)
Incubate cells with labeled CypB-CsA complex at 4°C (binding) or 37°C (internalization)
Analyze using:
Flow cytometry for quantitative assessment
Confocal microscopy for localization studies
Scintillation counting for radiolabeled components
Key parameters to measure:
Binding kinetics (Kd and number of binding sites)
Internalization rate of the complex
Differential accumulation of CypB versus CsA
Receptor recycling dynamics
Effect on T-cell activation using CD3-stimulated proliferation assays
Research has demonstrated that both free CypB and the CsA-CypB complex bind to T-lymphocyte surfaces with similar affinity (Kd values), but the complex shows different internalization dynamics with preferential accumulation of CsA within cells . This methodology allows detailed investigation of how CypB may enhance the immunosuppressive effects of CsA through targeted drug delivery.
Recent evidence demonstrating that CypB knockout mice are protected from NASH development opens significant research opportunities . To further elucidate CypB's role in NASH pathogenesis, researchers should consider these methodological approaches:
In vivo models and tissue-specific manipulations:
Conditional knockout strategies:
Generate hepatocyte-specific Ppib knockout using Alb-Cre
Create stellate cell-specific deletion using GFAP-Cre
Develop macrophage-specific deletion using LysM-Cre to assess inflammatory contributions
CypB inhibitor studies:
Test pan-cyclophilin inhibitors (e.g., CRV431/reconfilstat) versus selective CypB inhibitors
Establish dose-response relationships and treatment windows
Measure histological outcomes, inflammatory markers, and fibrosis progression
Mechanistic investigations:
ER stress pathway analysis:
Evaluate UPR markers (PERK, IRE1α, ATF6) in CypB-deficient versus wild-type hepatocytes
Measure ER calcium homeostasis using Fura-2 or genetically encoded calcium indicators
Assess mitochondrial function and ROS production
Multi-omics approaches:
Perform RNA-seq to identify transcriptional networks affected by CypB deletion
Use proteomics to identify CypB client proteins in hepatocytes
Apply lipidomics to characterize lipid profile changes in CypB-deficient livers
Cell-specific contributions:
Analyze hepatocyte lipid accumulation (Oil Red O staining)
Measure stellate cell activation markers (α-SMA, collagen)
Assess inflammatory cell recruitment and polarization
The experimental design should compare wild-type, CypA knockout, and CypB knockout mice under standard diet, western diet, CCl₄ treatment, or combined challenges to fully understand the specificity of CypB's contribution to NASH pathogenesis .
The discovery that CypB serum levels fluctuate across the menstrual cycle suggests important connections between CypB and hormonal regulation that warrant further investigation . A comprehensive research approach should include:
In vitro hormone response studies:
Cell culture models:
Treat primary human cells or relevant cell lines with physiological concentrations of estradiol, progesterone, FSH, and LH
Measure CypB gene expression, protein levels, and secretion
Analyze promoter activity using luciferase reporter assays to identify hormone-responsive elements
Receptor antagonist experiments:
Apply specific estrogen and progesterone receptor antagonists
Use siRNA knockdown of specific hormone receptors
Identify receptor subtypes mediating CypB regulation
In vivo models for hormonal manipulation:
Ovariectomized rodent models with hormone replacement:
Surgically remove ovaries to eliminate endogenous hormones
Administer controlled hormone replacement regimens
Measure serum and tissue CypB levels under different hormonal conditions
Transgenic approaches:
Generate CypB reporter mice to visualize expression patterns
Create conditional CypB knockout in hormone-responsive tissues
Assess reproductive phenotypes in CypB-deficient animals
Clinical studies with enhanced controls:
Design studies accounting for:
Precise cycle phase determination using hormone measurements
Oral contraceptive use and other medications
Age, BMI, and other potential confounders
Longitudinal sampling across multiple cycles to assess:
Consistency of CypB fluctuations
Correlation with specific hormone peaks
Relationship to cycle-dependent inflammatory markers
The demonstrated correlation between CypB, LH (r=0.743, p=0.009 at periovulatory phase), and FSH (r=0.633, p=0.036 at mid-luteal phase) provides a foundation for investigating potential regulatory relationships and functional significance in reproductive physiology .
The recombinant human Cyclophilin-B protein with a His tag is typically expressed in HEK293 cells or Escherichia coli. The His tag is a polyhistidine sequence added to the protein to facilitate purification via metal affinity chromatography . The recombinant protein consists of 190 amino acids and has a molecular weight of approximately 22 kDa .
Cyclophilin-B is both secreted and retained in the endoplasmic reticulum (ER). It plays a crucial role in ER redox homeostasis and has proinflammatory properties. High levels of Cyclophilin-B are found in inflammatory processes such as sepsis and rheumatoid arthritis . Additionally, Cyclophilin-B mediates chemotaxis and T cell adhesion to fibronectin .
The production of recombinant Cyclophilin-B involves the following steps:
Recombinant Cyclophilin-B is used in various research areas, including: