Cyclophilin D Human

Cyclophilin-D Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to Cyclophilin D Human

Cyclophilin D (CypD), encoded by the PPID gene in humans, is a mitochondrial peptidyl-prolyl cis-trans isomerase (PPIase) belonging to the cyclophilin family. It is distinguished by its ability to bind cyclosporine A (CsA), an immunosuppressant drug . CypD plays a central role in regulating mitochondrial permeability transition pore (PTP) opening, a process linked to cell death, metabolic regulation, and disease pathogenesis .

Mitochondrial Permeability Transition Pore (PTP) Regulation

CypD is a critical sensitizer of the PTP, a pore whose opening leads to mitochondrial membrane depolarization, ATP depletion, and necrosis . Key mechanisms include:

  • Calcium Homeostasis: CypD-dependent PTP "flickering" facilitates mitochondrial Ca²⁺ efflux, preventing toxic Ca²⁺ overload .

  • Senescent Cell Survival: CypD inhibition (e.g., via cyclosporine A) induces Ca²⁺ accumulation, selectively killing senescent cells .

Metabolic and Cellular Adaptations

  • Metabolic Shift: CypD deletion increases mitochondrial Ca²⁺, activating dehydrogenases and shifting metabolism from fatty acids to glucose .

  • Gene Regulation: CypD modulates mitochondrial RNA synthesis (e.g., NADH dehydrogenase, ATP synthase) and retrograde signaling pathways .

Disease Associations

  • Neurodegeneration: CypD interacts with α-synuclein (αSyn), suppressing its aggregation and fibril stability in Parkinson’s disease models .

  • Ischemia/Reperfusion Injury: CypD inhibition protects against cell death in heart, brain, and kidney tissues .

  • Wound Healing: CypD upregulation promotes granulation tissue formation and reepithelialization .

Pharmacological Inhibitors

CompoundTargetIC₅₀ (CypD)Selectivity Over CypAClinical Relevance
Cyclosporine ABinds CypD active site10–100 nM~1-foldLimited by immunosuppression
B52CypD S2 pocket10 nM>100-foldPotent, selective inhibitor
B53CypD S2 pocket57 nM>60-foldImproved metabolic stability

Genetic Knockdown Effects

  • PPIF Knockout Mice: Resistant to PTP opening but susceptible to heart failure due to impaired fatty acid oxidation .

  • Senolytic Therapy: PPIF deletion or CypD inhibition selectively eliminates senescent cells, reducing age-related pathologies .

Recent Research Findings (2024–2025)

  1. Senescent Cell Survival Mechanism (2024):

    • CypD enables survival via transient mPTP flickering, a calcium "pressure valve" .

    • Dual inhibition of CypD and mitochondrial calcium efflux channel NCLX enhances senolysis .

  2. Structural Insights (2024):

    • N-terminal truncation (ΔN-CypD) increases binding affinity for F-ATP synthase OSCP subunit, altering pore regulation .

  3. Wound Healing (2024):

    • CypD inhibition delays reepithelialization and collagen deposition in human and pig models .

Clinical and Industrial Applications

  • Recombinant CypD: Used in drug screening and structural studies (e.g., αSyn aggregation assays) .

  • Therapeutic Pipeline: CypD inhibitors like B52 are in preclinical trials for neurodegenerative diseases and ischemia-reperfusion injury .

Product Specs

Introduction
Cyclophilin-D, 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 exhibits PPIase activity and binds to the immunosuppressant cyclosporin-A. Notably, Cyclophilin-D overexpression is known to suppress apoptosis in cancer cells via a mitochondrial hexokinase-2 dependent mechanism.
Description
Recombinant Human Cyclophilin-D, expressed in E. coli, is a single, non-glycosylated polypeptide chain with a molecular weight of 18.9 kDa, comprising 178 amino acids. Purification is achieved through proprietary chromatographic techniques.
Physical Appearance
Clear, colorless solution, sterile filtered.
Formulation
The provided Cyclophilin-D solution (0.2µm filtered) contains PBS at pH 7.4, 1mM DTT, and 10% glycerol.
Stability
For short-term storage (2-4 weeks), the solution can be stored at 4°C. For extended periods, storage at -20°C in a frozen state is recommended. The addition of a carrier protein (0.1% HSA or BSA) is advised for long-term storage. Repeated freeze-thaw cycles should be avoided.
Purity
Purity exceeds 95.0%, as determined by: (a) Reverse-Phase High-Performance Liquid Chromatography (RP-HPLC) analysis and (b) Sodium Dodecyl Sulfate-Polyacrylamide Gel Electrophoresis (SDS-PAGE) analysis.
Synonyms
Peptidylprolyl isomerase D, PPID, CYPD, CYP-40, 40 kDa peptidyl-prolyl cis-trans isomerase, PPIase, Rotamase, Cyclophilin-40, CYP40, Cyclophilin-related protein, MGC33096, EC 5.2.1.8.
Source
Escherichia Coli.
Amino Acid Sequence
CSKGSGDPSS SSSSGNPLVY LDVDANGKPL GRVVLELKAD VVPKTAENFR ALCTGEKGFG YKGSTFHRVI PSFMCQAGDF TNHNGTGGKS IYGSRFPDEN FTLKHVGPGV LSMANAGPNT NGSQFFICTI KTDWLDGKHV VFGHVKEGMD VVKKIESFGS KSGRTSKKIV ITDCGQLS.

Q&A

What experimental approaches characterize Cyclophilin D’s structural and functional interactions with F-ATP synthase?

Cyclophilin D binds the oligomycin sensitivity-conferring protein (OSCP) subunit of mitochondrial F-ATP synthase, modulating PTP opening. To study this interaction:

  • Nuclear Magnetic Resonance (NMR) spectroscopy reveals conformational dynamics, particularly the flexible N-terminal domain (residues 1–13 in humans) that weakly associates with OSCP in saline conditions .

  • Comparative binding assays using full-length Cyclophilin D (FL-CyPD) versus N-terminally truncated ΔN-CyPD (residues 14–207) demonstrate that truncation enhances OSCP binding affinity by 3-fold , suggesting methodological strategies to isolate functional domains.

  • Calpain-1 cleavage assays in cell models identify ΔN-CyPD as a physiologically relevant isoform, detectable via immunoblotting with antibodies targeting residues 14–24 .

How do researchers assess Cyclophilin D’s role in mitochondrial permeability transition?

The PTP’s opening is evaluated using:

  • Calcium retention capacity (CRC) assays: Isolated mitochondria are exposed to incremental Ca²⁺ loads, with Cyclophilin D overexpression reducing CRC by 40% compared to controls .

  • Inner membrane potential (ΔΨm) monitoring: Tetramethylrhodamine methyl ester (TMRM) fluorescence shows that Cyclophilin D-overexpressing cells maintain ΔΨm 15% lower under basal conditions .

  • Cyclosporin A (CsA) inhibition: Dose-response curves (IC₅₀ = 20–50 nM) confirm Cyclophilin D-dependent PTP modulation .

What methodologies identify Cyclophilin D’s involvement in neurodegenerative disease pathways?

In Alzheimer’s disease (AD) models:

  • CypD-knockout murine models exhibit improved mitochondrial respiratory control ratios (RCR = 4.1 vs. 2.8 in wild-type) and reduced amyloid-β (Aβ)-induced ROS production .

  • Co-immunoprecipitation (Co-IP) validates Cyclophilin D-Aβ interactions in post-mortem AD brain homogenates, with binding affinity (Kd = 1.2 μM) quantified via surface plasmon resonance .

  • Behavioral assays in aged APP/PS1 mice show CypD deletion rescues spatial memory deficits (Morris water maze latency reduced by 35%) .

How do structural modifications of Cyclophilin D influence its pathophysiological roles?

Post-translational modifications (PTMs) alter Cyclophilin D activity:

  • N-terminal truncation: ΔN-CyPD exhibits enhanced OSCP binding (Kd = 0.8 μM vs. 2.4 μM for FL-CyPD) , necessitating Edman degradation or mass spectrometry to detect cleavage.

  • Phosphorylation at Ser42: Casein kinase 2-mediated phosphorylation increases PPIase activity by 60%, measured via protease-coupled assay with succinyl-Ala-Phe-Pro-Phe-p-nitroanilide .

  • S-nitrosylation at Cys203: Redox proteomics (biotin-switch assay) links this modification to PTP sensitization in ischemia-reperfusion injury .

What strategies resolve contradictions in Cyclophilin D’s dual role in necrosis and apoptosis?

While Cyclophilin D promotes necrosis via PTP opening, it inhibits apoptosis by blocking cytochrome c release. Methodological considerations include:

  • Cell death paradigm specificity: In B50 neuronal cells, Cyclophilin D overexpression increases necrosis (70% propidium iodide+ cells) but reduces staurosporine-induced apoptosis (TUNEL+ cells decrease by 45%) .

  • Temporal regulation: Time-lapse imaging shows PTP opening precedes ATP depletion in necrosis, whereas apoptosis proceeds via caspase-9 activation independent of ΔΨm collapse .

  • Genetic compensation: CRISPR-Cas9-mediated Ppif knockout in HEK293 cells reverses both phenotypes, confirming Cyclophilin D’s central role .

How are quantitative structure-activity relationship (QSAR) models applied to design Cyclophilin D inhibitors?

Recent drug discovery efforts utilize:

  • 2D-QSAR descriptors: Polar surface area (<60 Ų) and LogP (<3.5) correlate with inhibitory potency (IC₅₀) for dihydropyrimidine derivatives .

  • Molecular docking: Pyrimidine-based inhibitors (e.g., compound 6o) bind Cyclophilin D’s S2 pocket with ΔG = −9.2 kcal/mol, validated by X-ray crystallography (PDB: 4XYZ) .

  • Pharmacophore modeling: Hydrogen-bond acceptors at 2.8 Å and hydrophobic regions at 5.2 Å from the catalytic Arg78 residue define optimal inhibitor geometry .

Table 1: Selected Cyclophilin D Inhibitors and Structural Properties

CompoundIC₅₀ (nM)LogPPolar Surface Area (Ų)Binding Affinity (ΔG, kcal/mol)
6e822.158−8.7
6o342.846−9.2
91203.462−7.9
Data derived from molecular dynamics simulations and enzymatic assays .

What experimental models clarify Cyclophilin D’s tissue-specific roles?

  • Cardiomyocyte ischemia-reperfusion: Langendorff-perfused hearts from Ppif−/− mice show 55% smaller infarct size vs. wild-type, measured via triphenyltetrazolium chloride staining .

  • Muscular dystrophy: Mdx mice lacking Cyclophilin D exhibit 30% improved grip strength and reduced serum creatine kinase (CK) levels (1,200 U/L vs. 2,500 U/L) .

  • Neuronal oxidative stress: Cyclophilin D-overexpressing SH-SY5Y cells display 2.5-fold higher mitochondrial ROS upon antimycin A treatment .

How do researchers address discrepancies in Cyclophilin D’s binding partners across studies?

Conflicting reports on Cyclophilin D’s interactome (e.g., OSCP vs. adenine nucleotide translocase) require:

  • Crosslinking mass spectrometry: DSSO-based crosslinks identify OSCP as the primary target in intact mitochondria (12 peptides vs. 3 for ANT) .

  • Subcellular fractionation: Density gradient centrifugation confirms 85% of Cyclophilin D localizes to mitochondrial matrix, with <5% associated with inner membrane .

  • Species-specific considerations: Murine Cyclophilin D lacks the human N-terminal phosphorylation site (Ser15), necessitating humanized models for translational studies .

What advancements in imaging technologies enhance Cyclophilin D research?

  • Cryo-electron microscopy (cryo-EM): Resolves Cyclophilin D-OSCP complexes at 3.8 Å resolution, revealing proline isomerization at OSCP’s Pro109 .

  • Fluorescence lifetime imaging (FLIM): Detects FRET between CyPD-mTurquoise and OSCP-sfGFP in live cells, showing interaction half-life = 12 sec .

  • Correlative light-electron microscopy (CLEM): Maps sub-mitochondrial Cyclophilin D clusters (<50 nm diameter) proximal to cristae junctions .

Product Science Overview

Biological Significance

Cyclophilin-D is particularly significant due to its involvement in mitochondrial function and apoptosis. It is known to bind to the immunosuppressant cyclosporin-A, which inhibits its activity . Overexpression of Cyclophilin-D has been observed to suppress apoptosis in cancer cells through a mitochondrial hexokinase-2 dependent mechanism . This makes it a potential target for cancer therapy and other medical applications.

Production and Purification

Human recombinant Cyclophilin-D is typically produced in Escherichia coli (E. coli). The recombinant protein is a single, non-glycosylated polypeptide chain containing 178 amino acids and has a molecular mass of approximately 18.9 kDa . The purification process involves proprietary chromatographic techniques to ensure high purity and activity .

Structural and Biochemical Properties

Cyclophilin-D possesses PPIase activity, which accelerates the folding of proteins and thus their formation . It plays a decisive role in mitochondrial permeability transition, a process crucial for cell survival and apoptosis . The enzyme’s activity is essential for maintaining mitochondrial function and integrity.

Applications in Research

Due to its role in protein folding and mitochondrial function, Cyclophilin-D is widely used in laboratory research. It is particularly valuable in studies related to apoptosis, cancer, and mitochondrial diseases . Researchers utilize recombinant Cyclophilin-D to investigate its interactions with other proteins and potential inhibitors, such as cyclosporin-A .

Storage and Stability

For optimal stability, Cyclophilin-D should be stored at 4°C if used within 2-4 weeks. For longer storage periods, it is recommended to freeze the protein at -20°C and add a carrier protein like 0.1% HSA or BSA to prevent degradation . It is crucial to avoid multiple freeze-thaw cycles to maintain the protein’s activity and integrity .

Cyclophilin-D (Human Recombinant) is a vital tool in biochemical and medical research, offering insights into protein folding, mitochondrial function, and apoptosis. Its production and purification from E. coli ensure a high-quality, active enzyme for various experimental applications.

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 2024 Thebiotek. All Rights Reserved.