Cyclophilin A Human

Cyclophilin-A Human Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Structure and Biochemical Properties

Cyclophilin A is a cytosolic protein with a molecular weight of 18 kDa in its native form . Recombinant variants, such as those fused with a His tag, may exhibit slightly higher molecular weights (e.g., 20 kDa) . Its structure comprises:

  • An eight-stranded antiparallel β-barrel

  • Two α-helices flanking the barrel

  • A hydrophobic core critical for binding cyclosporin A (CsA)

Biological Functions

Cyclophilin A plays critical roles in:

  • Protein folding: Facilitates conformational changes via PPIase activity

  • Immune regulation: Binds CsA to inhibit calcineurin, suppressing T-cell activation

  • Viral infectivity: Incorporated into HIV-1 virions to enhance infectivity

  • Cellular signaling: Modulates oxidative stress and inflammation via interaction with CD147

Table 2: Disease Associations of Cyclophilin A

Disease CategoryMechanism and ImpactReference
Cardiovascular diseasesPromotes vascular remodeling, atherosclerosis, and heart failure via ROS production
HIV infectionBinds HIV-1 capsid proteins; essential for viral replication and infectivity
CancerOverexpressed in gastric, breast, and lung cancers; drives proliferation and metastasis
NeurodegenerationCo-localizes with apoptosis-inducing factor (AIF) in ALS and Alzheimer’s models
Liver injuryActs as a damage-associated molecular pattern (DAMP) in acetaminophen toxicity

Pharmacological Inhibitors

  • Cyclosporin A: Forms a complex with Cyclophilin A to inhibit calcineurin and mitochondrial permeability transition .

  • Non-immunosuppressive analogs: Target extracellular Cyclophilin A in inflammatory diseases .

Clinical Applications

  • Anti-eCyPA antibodies: Neutralize extracellular Cyclophilin A to mitigate cardiac hypertrophy and myocardial stiffening .

  • siRNA/shRNA: Reduces tumor growth in xenograft models of non-small-cell lung cancer .

Recombinant Production and Research Tools

Recombinant Cyclophilin A (e.g., ab86219, 3589-CAB) is produced in E. coli with:

  • Purity: >95% (SDS-PAGE verified)

  • Activity: >650 nmol/min/mg (PPIase assay)

  • Applications: Protein interaction studies, drug screening, and viral pathogenesis research

Clinical Correlations

  • Plasma Cyclophilin A levels correlate with disease severity in heart failure and inflammatory conditions .

  • Urinary Cyclophilin A is elevated in acetaminophen-induced liver injury .

Future Directions

Further research is needed to:

  • Identify novel receptors beyond CD147 for extracellular Cyclophilin A .

  • Develop isoform-specific inhibitors to minimize off-target effects .

Product Specs

Introduction
Peptidyl-prolyl cis-trans isomerases (PPIases) like Cyclophilin-A play a crucial role in protein folding by catalyzing the cis-trans isomerization of proline imidic peptide bonds. This protein is known to bind cyclosporin A, potentially contributing to its immunosuppressive effects. Additionally, Cyclophilin-A interacts with several HIV proteins, including p55 gag, Vpr, and capsid protein, and is essential for the formation of infectious HIV virions. The human genome contains multiple pseudogenes of Cyclophilin-A located on different chromosomes.
Description
Recombinant Human Cyclophilin-A, expressed in E. coli, is a non-glycosylated polypeptide with a 20 amino acid His tag at its N-terminus. This protein consists of 185 amino acids (including the His tag; the native protein sequence comprises residues 1-165) and has a molecular weight of 20 kDa. Purification is achieved through proprietary chromatographic techniques, resulting in a single polypeptide chain.
Physical Appearance
A clear, colorless solution free from particulate matter.
Formulation
The protein is supplied as a 1 mg/ml solution in a buffer consisting of 20mM Tris-HCl (pH 8.0), 20mM NaCl, 0.5mM DTT, and 10% glycerol.
Stability
For short-term storage (up to 4 weeks), the product can be stored at 4°C. For extended storage, freeze the solution at -20°C. The addition of a carrier protein like HSA or BSA (0.1%) is recommended for long-term storage. To maintain product integrity, avoid repeated freeze-thaw cycles.
Biological Activity
The enzyme exhibits a specific activity exceeding 650 nmol/min/mg. This is determined by measuring the enzyme's ability to cleave 1 nanomole of the substrate suc-AAFP-PNA per minute at 37°C in a Tris-HCl buffer (pH 8.0) using chymotrypsin.
Purity
SDS-PAGE analysis indicates a purity level greater than 95%.
Synonyms
Peptidylprolyl isomerase A, CYPA, CYPH, MGC12404, MGC23397, MGC117158, PPIase A, Rotamase A, PPIA, Peptidyl-prolyl cis-trans isomerase A, EC 5.2.1.8, Cyclophilin A, Cyclosporin A-binding protein.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MVNPTVFFDI AVDGEPLGRV SFELFADKVP KTAENFRALSTGEKGFGYKG SCFHRIIPGF MCQGGDFTRH NGTGGKSIYG EKFEDENFIL KHTGPGILSMANAGPNTNGS QFFICTAKTE WLDGKHVVFG KVKEGMNIVE AMERFGSRNG KTSKKITIADCGQLE.

Q&A

What is the molecular structure of human Cyclophilin A and how does it relate to its function?

Human Cyclophilin A features an eight-stranded antiparallel β-barrel structure, as revealed by X-ray crystallography . This structure contains a hydrophobic pocket that serves as both the binding site for cyclosporin A and the catalytic site for its peptidyl-prolyl isomerase activity. The coincidence of these sites explains how cyclosporin A inhibits CypA's enzymatic function . For experimental investigations, researchers typically use E. coli-derived recombinant human Cyclophilin A spanning amino acids Met1-Glu165 (accession # P62937) . This structural arrangement provides the foundation for CypA's role in protein folding, trafficking, and immunological functions.

How does Cyclophilin A differ from other members of the cyclophilin family?

While human cyclophilins consist of 16 structurally distinct family members, CypA stands out as the most abundant, constituting approximately 0.1-0.6% of total cytosolic proteins . Unlike Cyclophilin B, which was traditionally considered the secreted isoform, CypA can unexpectedly be secreted in response to inflammatory stimuli despite being primarily cytoplasmic . This distinction is particularly evident in rheumatoid arthritis patients, where CypA has been detected in synovial fluids through immunoblotting and N-terminal sequencing . When developing detection methods, researchers should note that some antibodies show cross-reactivity with other cyclophilins, particularly Cyclophilin E (approximately 50% cross-reactivity in direct ELISAs) .

What enzymatic activities does Cyclophilin A possess?

The primary enzymatic function of Cyclophilin A is its peptidyl prolyl cis-trans isomerase (PPIase) activity, which catalyzes the isomerization of peptide bonds from trans form to cis form at proline residues . This activity is crucial for protein folding as it accelerates the rate-limiting step in the folding process for proline-containing proteins. Experimentally, this activity can be measured using spectrophotometric assays with specific peptide substrates containing proline residues . The PPIase activity is inhibited by cyclosporin A, which binds competitively to the same hydrophobic pocket that serves as the catalytic site . Recent research indicates that CypA's enzymatic function extends beyond simple protein folding to include roles in signal transduction and T-cell activation .

What are the optimal techniques for detecting Cyclophilin A in experimental samples?

For detecting human Cyclophilin A in experimental samples, Western blot analysis using specific antibodies provides reliable results. Studies have successfully used rat anti-human Cyclophilin A monoclonal antibodies to detect a specific band at approximately 18 kDa in various cell lines including MOLT-4 human acute lymphoblastic leukemia cells, HeLa cells, and PANC-1 human pancreatic carcinoma cells . For tissue localization, immunofluorescence techniques can visualize CypA in both nuclei and cytoplasm using fluorescent-conjugated secondary antibodies . Automated capillary-based detection systems like Simple Western have also proven effective, detecting CypA at approximately 22 kDa in cell lysates . When designing detection protocols, researchers should optimize antibody dilutions (typically starting at 0.1 μg/mL for Western blots) and consider potential cross-reactivity with other cyclophilin family members .

How can Cyclophilin A activity be measured quantitatively in biological samples?

Quantitative measurement of Cyclophilin A activity in biological samples generally relies on assessing its PPIase activity through:

  • Spectrophotometric assays using peptide substrates with proline residues, where cis-trans isomerization induces measurable changes in absorbance or fluorescence

  • Protease-coupled assays where proteolysis rates depend on CypA-catalyzed isomerization

  • NMR-based methods that directly observe conformational changes

For validating specificity, researchers should perform parallel assays in the presence of cyclosporin A, which inhibits CypA by binding to its hydrophobic pocket and competitively blocking the PPIase activity . Studies of staphylococcal cyclophilin have shown that the CypA-CsA complex exhibits significantly increased thermodynamic stability compared to unbound CypA, suggesting structural stabilization upon inhibitor binding . This property can be exploited in thermal shift assays as an alternative approach to measuring binding interactions.

What purification strategies yield the highest quality Cyclophilin A for in vitro studies?

For high-quality Cyclophilin A preparation, recombinant expression in E. coli systems has proven effective . The purification protocol typically involves:

  • Bacterial expression of the full-length human CypA (Met1-Glu165)

  • Cell lysis under controlled conditions

  • Sequential chromatography steps:

    • Affinity chromatography (often using cyclosporin A-based matrices)

    • Ion exchange chromatography

    • Size exclusion chromatography to achieve high purity

The final preparation can be supplied either lyophilized or as a 0.2 μm filtered solution in PBS depending on experimental requirements . Quality control should include SDS-PAGE, enzymatic activity assays, and mass spectrometry to confirm identity and purity. For studies involving the CypA-CsA complex, cyclosporin A binding activity is preserved even in partially unfolded intermediates, as demonstrated in unfolding studies with urea . This resilience indicates that properly purified CypA maintains its functional properties under various experimental conditions.

How does Cyclophilin A contribute to the pathogenesis of inflammatory diseases?

Cyclophilin A plays multiple roles in inflammatory disease pathogenesis, particularly in rheumatoid arthritis where it has been detected in synovial fluids of patients . The concentration of CypA in synovial fluids significantly correlates with the number of neutrophils in the cellular infiltrate and is elevated in more acute cases of joint swelling . Mechanistically, CypA acts as a proinflammatory mediator through several pathways:

  • It functions as a chemotactic factor for polymorphonuclear leukocytes and monocytes, recruiting inflammatory cells to sites of inflammation

  • It binds to the surface of T cells, potentially modulating immune responses

  • It is secreted during inflammatory conditions as a proinflammatory product

The effectiveness of cyclosporin A in rheumatoid arthritis may be partially explained by its inhibition of CypA, suggesting that CypA contributes to disease progression beyond serving as a simple biomarker . Research approaches should consider both the intracellular and extracellular functions of CypA when investigating inflammatory mechanisms.

What molecular mechanisms explain Cyclophilin A's role in HIV-1 infection?

Cyclophilin A is essential for effective HIV-1 replication through distinct mechanisms at both entry and post-entry stages of the viral life cycle . At the molecular level:

  • CypA is specifically incorporated into nascent HIV-1 virions by binding to the capsid (CA) region of the Gag precursor protein

  • The hydrophobic pocket of CypA interacts with an exposed loop of the HIV-1 capsid protein

  • This interaction is critical for viral infectivity, as demonstrated by experiments where CypA incorporation is prevented either by mutations in the CypA packaging signal of CA or by the presence of cyclosporin A

Viruses lacking CypA exhibit significantly reduced infectivity compared to wild-type viruses . The competitive inhibition of CypA-CA interactions by cyclosporin A explains why CsA can hamper HIV-1 replication . For research methodologies, this mechanism suggests that targeting the CypA-CA interaction interface could provide a strategy for developing novel antiretroviral agents with a high barrier to resistance.

How is Cyclophilin A implicated in cancer progression and metastasis?

Cyclophilin A is generally overexpressed in cancer and regulates multiple aspects of malignant transformation and metastasis . The mechanisms underlying CypA's oncogenic roles include:

  • Modulation of cell proliferation pathways through its PPIase activity and interactions with signaling molecules

  • Enhancement of cell survival by influencing apoptotic pathways

  • Promotion of angiogenesis, tissue invasion, and metastatic spread

  • Involvement in therapy resistance mechanisms

High CypA expression correlates with poor outcomes in patients with various cancer types, positioning it as both a potential biomarker and therapeutic target . Experimentally, cancer researchers can study CypA's role using knockdown/knockout approaches in cancer cell lines and animal models, followed by functional assays for proliferation, migration, invasion, and in vivo tumor growth. When designing such studies, attention should be paid to both the intracellular signaling functions of CypA and its potential paracrine effects when secreted into the tumor microenvironment.

How should researchers control for specificity when studying Cyclophilin A among other cyclophilins?

When investigating Cyclophilin A specifically among other cyclophilin family members, researchers should implement multiple controls for specificity:

  • Antibody selection: Choose antibodies validated for minimal cross-reactivity with other cyclophilins. Some anti-CypA antibodies show approximately 50% cross-reactivity with Cyclophilin E and less than 15% with other cyclophilins in direct ELISAs

  • Molecular detection: Western blotting can distinguish CypA (~18 kDa) from other cyclophilins based on molecular weight differences . Confirmatory techniques such as mass spectrometry or N-terminal sequencing can provide definitive identification, as demonstrated in studies of synovial fluid where an ~18-kD protein was confirmed as CypA through sequencing

  • Functional validation: Compare PPIase activity in the presence of specific inhibitors or with recombinant protein controls

  • Genetic approaches: Use siRNA or CRISPR-Cas9 with sequences specific to CypA while monitoring expression of other cyclophilins to confirm selective targeting

For experiments involving cyclosporin A, which can bind multiple cyclophilins, researchers should design controls that distinguish effects mediated specifically through CypA versus other cyclophilin family members.

What are the critical parameters for assessing Cyclophilin A toxicity in experimental systems?

Assessment of Cyclophilin A toxicity requires careful experimental design considering multiple parameters:

  • Dose-response relationships: Studies with recombinant human CypA have shown that toxicity becomes evident at high doses (750 mg/kg) when administered intraperitoneally or subcutaneously to mice

  • Route of administration: Toxicity profiles differ between intraperitoneal and subcutaneous administration routes

  • Sex differences: Female and male mice show different susceptibility to CypA toxicity, with manifestations including kidney injury and altered granulocyte/lymphocyte ratios in blood

  • Molecular markers: Expression of acute phase protein genes (Saa1, Saa2) is induced at doses as low as 0.1-2 mg/mouse, while maximal doses (750 mg/kg) significantly stimulate expression of multiple acute phase protein genes

  • Organ-specific effects: Prioritize assessment of kidney function, as renal injury appears to be a primary manifestation of CypA toxicity

When designing toxicity studies, researchers should include time-course analyses to distinguish between acute and chronic effects, as well as comprehensive hematological and biochemical panels to detect systemic responses.

How can researchers effectively study the structural changes in Cyclophilin A upon binding to cyclosporin A?

To study structural changes in Cyclophilin A upon cyclosporin A binding, researchers should employ complementary structural biology approaches:

  • X-ray crystallography: This has successfully revealed the structure of human recombinant cyclophilin complexed with cyclosporin A, showing that the binding occurs at a hydrophobic pocket that coincides with the prolyl isomerase substrate-binding site

  • Nuclear magnetic resonance (NMR) spectroscopy: This technique has identified the specific binding site for cyclosporin A and can provide dynamic information about conformational changes

  • Unfolding studies: Research with staphylococcal cyclophilin has demonstrated that urea-induced unfolding of both unbound and CsA-bound CypA is completely reversible and proceeds via stable intermediates . The thermodynamic stability of CypA increases significantly in the presence of CsA, indicating structural stabilization upon binding

  • Functional assays: Even in partially unfolded intermediate states, CypA maintains its CsA binding activity, suggesting that the binding interface remains structurally preserved during partial denaturation

For comprehensive structural analysis, researchers should combine these approaches to capture both static structures and dynamic conformational changes induced by CsA binding.

How can Cyclophilin A be utilized as a target for developing novel therapeutic agents?

Developing novel therapeutic agents targeting Cyclophilin A represents a promising approach for multiple disease conditions:

  • For inflammatory diseases: CypA inhibitors could potentially reduce inflammatory cell recruitment and activation. Research has shown that CypA is present in synovial fluids of rheumatoid arthritis patients and correlates with neutrophil infiltration, suggesting that targeted inhibition could ameliorate inflammation

  • For viral infections: Since CypA is necessary for HIV-1 replication through its interaction with the viral capsid protein, compounds disrupting this interaction without affecting normal CypA functions could serve as antiretroviral agents with a high barrier to resistance

  • For cardiovascular diseases: CypA acts as a critical mediator in cardiovascular pathologies, indicating that targeted inhibition could potentially reduce disease progression

  • Drug development approaches:

    • Structure-based design utilizing the X-ray crystal structure of human cyclophilin and its binding site for cyclosporin A

    • Non-immunosuppressive CsA derivatives that retain CypA binding but lack calcineurin inhibition

    • Novel scaffolds identified through high-throughput screening against the PPIase activity

When designing CypA-targeting compounds, researchers should aim to develop agents that block specific pathological functions of CypA while minimizing effects on its physiological roles and without affecting other cyclophilin isoforms .

What approaches can resolve contradictory findings about Cyclophilin A's role in different disease contexts?

Resolving contradictory findings about Cyclophilin A's roles across different disease contexts requires systematic methodological approaches:

  • Context-specific analysis: CypA functions differently depending on cellular location (intracellular vs. secreted) and disease context. For example, while generally considered proinflammatory, CypA may have protective roles in certain conditions

  • Standardized experimental systems:

    • Use consistent cell types and experimental conditions when comparing CypA functions

    • Report detailed experimental parameters including CypA concentrations, incubation times, and cell activation states

    • Specify the exact form of CypA used (full-length vs. truncated, post-translational modifications)

  • Molecular mechanism dissection:

    • Distinguish between enzymatic (PPIase) dependent and independent functions

    • Identify specific binding partners in each disease context

    • Use point mutations that selectively affect different CypA functions

  • In vivo validation with appropriate disease models:

    • Use conditional knockout approaches for tissue-specific CypA deletion

    • Consider timing of CypA manipulation relative to disease progression

    • Account for potential compensatory mechanisms involving other cyclophilins

  • Meta-analysis approaches: Systematically compare findings across multiple studies while accounting for methodological differences to identify consistent patterns despite apparent contradictions.

What are the most promising applications of Cyclophilin A in biomarker development?

Cyclophilin A shows significant potential as a biomarker across multiple disease contexts:

  • For inflammatory diseases: CypA concentrations in synovial fluids correlate with neutrophil infiltration and acute joint swelling in rheumatoid arthritis patients, suggesting utility as a biomarker of disease activity

  • For cardiovascular diseases: CypA is a critical mediator in cardiovascular pathologies, and its levels could potentially indicate disease progression or response to therapy

  • For viral infections: Since CypA is necessary for HIV-1 replication, monitoring CypA-virus interactions could provide insights into viral dynamics or therapeutic responses

  • For cancer: CypA is generally overexpressed in cancer, and high expression correlates with poor outcomes, positioning it as a potential prognostic biomarker

  • Biomarker development approaches:

    • Develop standardized ELISA or automated immunoassays for CypA quantification in biological fluids

    • Establish normal reference ranges across different populations

    • Conduct longitudinal studies correlating CypA levels with disease progression and treatment responses

    • Evaluate CypA in biomarker panels to improve diagnostic or prognostic accuracy

When developing CypA as a biomarker, researchers should validate findings across multiple patient cohorts and consider how CypA levels may be influenced by confounding factors such as age, as CypA expression increases with aging .

Product Science Overview

Functions and Roles

CypA has a broad spectrum of functions and exists in both intracellular and secreted forms . Intracellularly, it is present in nearly all cells, either in the cytosol or nucleus, and functions as a chaperone and housekeeping protein . It also acts as a ligand for cyclosporine A (CsA), an immunosuppressive drug .

Secreted CypA, on the other hand, plays several roles:

  • Proinflammatory Factor: It acts as a chemoattractant and immune regulator .
  • Antitumor Immunity: It has been identified as a factor in antitumor immunity .
  • Clinical Applications: Experimental data suggest potential applications of recombinant human CypA (rhCypA) in cancer immunotherapy, stimulation of tissue regeneration, treatment of brain pathologies, and as a supportive treatment for CsA-based therapies .
Pharmacokinetics

Recent studies have analyzed the pharmacokinetics of rhCypA in mouse models . These studies involved isotope-labeling rhCypA with 125I and injecting it intraperitoneally (i.p.) or subcutaneously (s/c) into mice . The results showed:

  • Rapid and Even Tissue-Organ Distribution: rhCypA exhibited rapid and even distribution across tissues and organs .
  • Highest Tropism to Kidneys: The highest accumulation was observed in the kidneys, the primary excretory organ .
  • Crossing the Blood-Brain Barrier: For the first time, rhCypA was shown to cross the blood-brain barrier and accumulate in the brain .
Historical Context

CypA was originally isolated from bovine thymocytes and identified as the cytosolic receptor for the immunosuppressive cyclic undecapeptide cyclosporin A . The formation of a CypA-CsA complex impairs the transcription of genes related to the immune response and prevents the proliferation of T cells .

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.