TPO Human

Thyroid Peroxidase Human Recombinant
Shipped with Ice Packs
In Stock

Description

Physical Properties and Conservation

While the predicted molecular mass of TPO is approximately 36.3 kDa (338 amino acids), the protein typically migrates as 75-85 kDa on SDS-PAGE due to extensive glycosylation . This property highlights the significant post-translational modifications that contribute to TPO's functional characteristics.

TPO proteins demonstrate remarkable evolutionary conservation, with human, canine, and mouse variants exhibiting 69-75% sequence identity at the amino acid level . This high degree of conservation across species underscores the fundamental biological importance of this growth factor in mammalian physiology.

Endogenous Production

Thrombopoietin is primarily produced by the liver and kidney tissues . As a critical growth factor, it circulates in the bloodstream to reach its target cells in the bone marrow, where it exerts its primary biological functions.

Recombinant Production and Formulation

Recombinant human TPO (rhTPO) is typically produced in genetically modified cell lines, with Chinese Hamster Ovary (CHO) cells being a common expression system . The resulting protein may be either:

  1. Non-glycosylated protein containing the 174 amino acids of the receptor binding domain, with a molecular mass of 18.6 kDa

  2. Full-length glycosylated protein similar to the native form

Commercial rhTPO is often provided as a sterile filtered white lyophilized powder, formulated in buffers such as 10 mM sodium phosphate (pH 8.0) . The stability of lyophilized TPO is excellent at -20°C, with reconstituted material typically prepared at 0.1 mg/ml in sterile water .

Receptor Binding and Signaling

TPO functions as the ligand for the receptor encoded by the c-Mpl proto-oncogene . It binds to the TPO receptor (CD110) on the surface of megakaryocytes and their precursors, initiating signaling cascades that promote cell proliferation, differentiation, and maturation .

Primary Physiological Functions

The primary biological activities of TPO include:

  • Stimulation of megakaryocyte maturation and differentiation

  • Promotion of platelet production

  • Regulation of hematopoietic stem cell survival and proliferation

TPO acts as a highly specific platelet stimulator by promoting the differentiation of bone marrow hematopoietic stem cells into megakaryocytes and stimulating their growth and maturation .

Biological Activity Measurements

The biological activity of rhTPO is typically determined through cell-based assays. Activity is often measured by dose-dependent stimulation of MO7e cells, with effective concentrations typically less than 1 ng/ml . Alternatively, activity can be assessed by proliferation of TF-1 cells, with ED50 values (effective dose for 50% maximal response) typically ranging from 0.5-5 ng/mL .

Therapeutic Applications

Recombinant human thrombopoietin has shown significant therapeutic potential in various clinical conditions characterized by thrombocytopenia, including:

  • Chemotherapy-induced thrombocytopenia

  • Idiopathic thrombocytopenic purpura (ITP)

  • Sepsis-associated thrombocytopenia

  • Pneumonia-associated thrombocytopenia

Clinical Study Results in Pneumonia Patients

A retrospective cohort study conducted at Nanfang Hospital in China assessed the efficacy of rhTPO in ICU patients with pneumonia and thrombocytopenia. The study included 327 patients (149 in the rhTPO group and 178 in the no-rhTPO group) and revealed several significant findings :

ParameterrhTPO GroupNo-rhTPO GroupP-value
Platelet count increase within 7 days99.21 ± 102.6132.08 ± 43.8770.000
Clinical recovery rate within 7 days (%)65.818.50.000
Decrease in hemorrhagic scores after 7 days2.81 ± 2.8561.16 ± 2.1230.000
Cessation of bleeding events (%)66.737.30.000
Red blood cell transfusions needed3.639 ± 4.6305.818 ± 6.8580.009

These results demonstrate significant improvements in platelet counts, reduced bleeding, and decreased transfusion requirements in patients receiving rhTPO .

Meta-Analysis of TPO Efficacy in Sepsis Patients

  • Significantly increased platelet counts [standardized mean difference (SMD), 2.61; 95% confidence interval (CI), 1.28–3.94; P < 0.001]

  • Decreased 28-day mortality [relative risk (RR), 0.66; 95% CI, 0.46–0.97; P=0.03]

  • Reduced transfusion volume of blood products

  • Decreased length of ICU stay

The same analysis also found that when compared to conventional antibiotic therapy combined with intravenous immunoglobulin, rhTPO was associated with improved platelet counts on day 7 of treatment (SMD, 0.86; 95% CI, 0.54–1.17; P < 0.001) and reduced transfusion volume of blood products .

Laboratory and Research Utilities

Human TPO finds extensive application in various research settings, including:

  • Cell culture applications requiring megakaryocyte differentiation

  • Hematopoietic stem cell expansion protocols

  • Development and testing of platelet production methods in vitro

  • Studies of thrombopoiesis and megakaryocyte biology

  • Investigation of signaling pathways in hematopoietic cells

Commercial preparations of TPO for research typically include carrier-free recombinant proteins suitable for functional assays, Western blotting, and ELISA applications .

Experimental Considerations

When utilizing TPO in experimental settings, several factors should be considered:

  • The biological activity may vary between different preparations and manufacturers

  • Glycosylated and non-glycosylated forms may have different potencies

  • For cell culture applications, appropriate co-factors or additional cytokines may be required

  • Dose-response relationships should be established for each experimental system

Future Perspectives and Ongoing Research

The remarkable efficacy of rhTPO in treating thrombocytopenia suggests significant potential for broader applications in hematology and critical care medicine. Ongoing research continues to explore:

  • Optimization of dosing regimens for different clinical scenarios

  • Combination therapies with other cytokines or growth factors

  • Development of improved formulations with enhanced stability or activity

  • Exploration of new indications beyond established applications

  • Long-term safety profiles in various patient populations

The identification of rhTPO administration as an independent indicator affecting platelet recovery rate in clinical studies suggests that further investigation into its mechanisms and applications could yield additional therapeutic benefits .

Product Specs

Introduction
Thyroid Peroxidase (TPO) is a major autoantigen in human autoimmune thyroid disease, previously known as the 'microsomal antigen'. This integral membrane glycoprotein, found only on the apical plasma membrane of follicular epithelial cells, consists of two identical subunits, each weighing approximately 100 kDa. TPO, a hemoprotein, is crucial for thyroid hormone biosynthesis. It catalyzes the iodination of tyrosyl residues and the coupling of iodotyrosyl residues in thyroglobulin (TG) to form precursors of thyroid hormones T4 and T3.
Description
Recombinant Human Thyroid Peroxidase, produced in SF9 cells, is a glycosylated polypeptide chain of 834 amino acids. It has a molecular mass of 92,872 Daltons (excluding glycosylation) and a total mass of 101 kDa, including glycosylation. The TPO is expressed with a -6xHis tag and purified using proprietary chromatographic techniques.
Formulation
TPO is supplied in a solution of 16mM HEPES buffer at pH 7.6, 160mM NaCl, 0.08mM KI, and 20% glycerol.
Coating Concentration
The recommended coating concentration for TPO is 0.15-0.375 µg/ml. The optimal concentration may vary depending on the ELISA plate type and coating buffer used. TPO is suitable for biotinylation and iodination.
Purity
The purity of TPO is greater than 95% as determined by Densitometric Analysis.
Stability
TPO should be stored at 4°C if the entire vial will be used within 2-4 weeks. For long-term storage, freeze TPO at -20°C. Repeated freezing and thawing should be avoided.
Synonyms
Thyroid peroxidase, EC 1.11.1.8, TPO, MSA, TPX.
Source
Sf9 insect cells.

Q&A

What is the domain organization of human thyroid peroxidase?

Human thyroid peroxidase (TPO) consists of four distinct domains: the N-terminal domain, the peroxidase domain (POD), the complement control protein (CCP)-like domain, and the epidermal growth factor-like domain. These domains are stabilized by a critical disulfide bond between Cys146 in the POD and Cys756 in the CCP domain, which constrains molecular flexibility . This arrangement positions the enzyme active site, containing the haem group and calcium binding site, on the opposite face from common antibody binding regions. Understanding this domain organization is essential for interpreting enzyme kinetics, antibody interactions, and designing domain-specific experimental approaches.

The mature human TPO protein is derived from a 353 amino acid precursor that undergoes proteolytic processing to yield the functional enzyme. Researchers should note that the relative positions of these domains are relatively fixed due to the disulfide bond linkage, which has significant implications for experimental design, particularly when producing recombinant fragments for structural or immunological studies .

How can I assess TPO enzymatic activity in purified preparations?

The standard approach for measuring human TPO enzymatic activity is the guaiacol oxidation assay. This spectrophotometric method provides quantitative assessment of peroxidase function. The protocol involves:

  • Prepare samples of different TPO concentrations diluted in appropriate buffer with 0.25 mg/mL bovine serum albumin (BSA)

  • Mix with 2 mL of guaiacol solution (43 mmol/L guaiacol, 10 mmol/L Tris-HCl pH 8.0, 0.25 mg/mL BSA)

  • Incubate at 37°C for 20 minutes

  • Add hydrogen peroxide (10 μL, 60 mmol/L)

  • Record absorbance at 470 nm at 15, 30, and 60 seconds intervals

This methodology allows for comparative activity assessment across different TPO preparations. For more sensitive detection, especially with limited sample quantities, researchers may consider alternative substrates like tetramethylbenzidine (TMB) which offers enhanced chromogenic responses. When comparing activities between preparations, standardization against a reference preparation is essential for meaningful cross-laboratory comparisons .

What are the key immunodominant regions of human TPO?

Human TPO contains two major immunodominant regions that are primary targets of autoantibodies in autoimmune thyroid diseases:

  • Immunodominant Region A (IDR-A): This epitope spans across both the MPO-like and CCP-like domains, making it relatively fragmented. Due to this fragmentation, conformational rearrangements are likely required for the region to form a compact epitope recognized by autoantibodies.

  • Immunodominant Region B (IDR-B): This epitope is located predominantly on the surface of the MPO-like domain. Its accessibility to autoantibodies varies depending on TPO's orientation relative to the thyroid follicular cell membrane .

These regions are particularly important for researchers developing diagnostic tests or studying autoimmune mechanisms. When designing experiments to map epitopes or assess autoantibody binding, researchers should consider that conformational rearrangements may be necessary for full engagement with autoantibodies, as IDR-B is located near the dimer interface in most structural models .

How can cryo-electron microscopy be optimized for human TPO structural studies?

Cryo-electron microscopy (cryo-EM) has proven invaluable for elucidating TPO structure, particularly when complexed with antibodies. For optimal results when applying this technique to TPO studies:

  • Sample preparation considerations:

    • Purify TPO to homogeneity (>95% purity by SDS-PAGE)

    • Complex with Fab fragments of relevant antibodies to increase particle asymmetry and improve orientation distribution

    • Concentrate samples to approximately 2 mg/mL for optimal particle density

  • Data collection parameters:

    • Use high-end microscopes (e.g., Titan Krios 300kV with Falcon3 detector in counting mode)

    • Apply magnification of approximately 96,000× (corresponding to pixel size of ~0.83 Å/pixel)

    • Use a total dose of 43-45 e/Ų fractionated over 40 frames

    • Set defocus range from -2.4 to -0.9 μm with incremental steps of 0.3 μm

    • Implement automated data acquisition using specialized software (e.g., EPU)

  • Data processing workflow:

    • Perform motion correction on movie frames

    • Estimate the contrast transfer function (CTF)

    • Select particles automatically with manual verification

    • Conduct 2D and 3D classifications

    • Apply refinement techniques to achieve highest possible resolution

This methodology has achieved resolutions of 3.4-3.9 Å for TPO-antibody complexes, allowing detailed visualization of domain arrangements and antibody-binding interfaces .

What are the contrasting models of TPO dimerization and membrane orientation?

Molecular modeling approaches have revealed two plausible but contrasting models of TPO dimerization and membrane orientation, each with distinct implications for research:

"Trans" Model:

  • IDR-B is positioned on the membrane-facing side of the MPO-like domain

  • Active site accessibility is limited by the membrane orientation

  • Autoantibody access to IDR-B would require significant conformational changes or dimer dissociation

  • This model suggests more complex autoantibody-binding mechanisms

"Cis" Model:

  • IDR-B clusters toward the surface of the MPO-like domain facing the thyroid follicular lumen

  • Higher solvent accessibility of immunodominant regions

  • Less epitope fragmentation across domains

  • This model is slightly favored based on autoantibody accessibility considerations

The C-terminus in both models contains features consistent with a coiled-coil dimerization motif that likely anchors the TPO dimer in the apical membrane of thyroid follicular cells. When designing experiments to investigate TPO dimerization or membrane interactions, researchers should consider both models and design experiments that could distinguish between them, such as crosslinking studies or accessibility assays .

Feature"Trans" Model"Cis" Model
IDR-B LocationMembrane-facing sideLumen-facing side
Epitope AccessibilityLimited, requires conformational changeHigher, more solvent-exposed
Active Site OrientationAway from lumenToward lumen
Compatibility with Autoantibody BindingLess favorableMore favorable
Domain Rearrangement RequirementsSignificantModerate

How can TPO-autoantibody interactions be assessed experimentally?

For researchers investigating TPO-autoantibody interactions, several methodological approaches are recommended:

  • Inhibition ELISA methodology:

    • Prepare dilutions of purified TPO (800 to 3.1 ng/mL in appropriate buffer)

    • Pre-incubate with pooled TPOAb-positive patient sera or controls (1 hour at room temperature)

    • Add the pre-incubated TPO/TPOAb mixture to rhTPO-coated ELISA plate wells

    • Incubate for 1 hour at room temperature with shaking

    • Detect bound antibodies using anti-human IgG-HRP conjugate

    • Develop using TMB substrate and measure absorbance at 450 nm and 405 nm

  • TPO-antibody complex preparation for structural studies:

    • Mix purified TPO with Fab fragments at a 1:1.2 molar ratio

    • Incubate overnight at 4°C to ensure complete complex formation

    • Purify the complex using size-exclusion chromatography

    • Verify complex formation by SDS-PAGE and analytical SEC

    • Assess retained enzymatic activity of the complex using guaiacol oxidation assay

  • Epitope mapping approaches:

    • Generate a panel of TPO mutants with alterations in predicted epitope regions

    • Express recombinant fragments corresponding to different TPO domains

    • Perform competition binding experiments between monoclonal antibodies

    • Use hydrogen-deuterium exchange mass spectrometry to identify antibody footprints on TPO

These methods provide complementary data on the specificity, affinity, and structural basis of TPO-autoantibody interactions, which are crucial for understanding autoimmune disease mechanisms and developing targeted diagnostic tools.

What expression systems are most effective for producing recombinant human TPO?

Several expression systems have been employed for human TPO production, each with distinct advantages and limitations:

  • Insect cell expression (Baculovirus):

    • System of choice for most structural studies

    • Spodoptera frugiperda (Sf21) cells provide proper glycosylation and folding

    • Yields functionally active enzyme with proper heme incorporation

    • Supports production of TPO spanning Ser22-Gly353

  • Mammalian cell expression:

    • CHO or HEK293 cells provide mammalian-type glycosylation

    • More physiologically relevant post-translational modifications

    • Lower yield compared to insect cells but potentially higher activity

    • Better for antibody recognition studies where glycosylation affects epitope structure

  • Bacterial expression of domains:

    • Suitable for individual domains (particularly the peroxidase domain)

    • Lacks glycosylation and may require refolding protocols

    • Higher yield but lower enzymatic activity

    • Useful for basic binding studies or antibody generation

When selecting an expression system, researchers should consider their specific experimental needs. For structural studies or enzymatic characterization, insect or mammalian systems are strongly preferred due to their ability to produce properly folded and post-translationally modified protein .

What purification strategies yield the most homogeneous TPO preparations?

Obtaining highly homogeneous TPO preparations is crucial for structural and functional studies. A recommended multi-step purification strategy includes:

  • Initial capture:

    • Affinity chromatography using anti-TPO antibodies or engineered affinity tags

    • Concanavalin A lectin chromatography to exploit glycosylation properties

    • Consider using detergent-containing buffers if purifying full-length TPO from membranes

  • Intermediate purification:

    • Ion exchange chromatography (IEX) to separate based on charge differences

    • Hydroxyapatite chromatography, which has proven effective for TPO purification

  • Polishing steps:

    • Size exclusion chromatography (SEC) to separate monomers, dimers, and higher-order aggregates

    • Removal of affinity tags if used (e.g., TEV protease cleavage)

  • Quality control assessments:

    • Purity assessment by SDS-PAGE (>95% recommended for structural studies)

    • Enzymatic activity using guaiacol oxidation assay

    • Mass spectrometry to confirm identity and intact mass

    • Dynamic light scattering to assess homogeneity and aggregation state

For researchers preparing TPO for cryo-EM studies, an additional concentration step to approximately 2 mg/mL is recommended, with careful monitoring to prevent aggregation. Buffer optimization through thermal stability assays can significantly improve sample quality and stability during purification and subsequent experiments .

How do TPO autoantibodies differ in various autoimmune thyroid diseases?

TPO autoantibodies (TPOAbs) show distinct characteristics across different autoimmune thyroid conditions, which has important implications for research design:

  • Hashimoto's thyroiditis:

    • TPOAbs predominantly target conformational epitopes spanning IDR-A and IDR-B

    • Higher affinity and complement-activating capacity

    • Predominantly IgG1 and IgG4 subclasses

    • Often co-exist with antibodies against other thyroid antigens

  • Graves' disease:

    • Lower titers of TPOAbs compared to Hashimoto's thyroiditis

    • Different epitope recognition patterns with less focus on IDR-B

    • Predominantly IgG1 subclass with less IgG4

    • Usually accompanied by TSH receptor stimulating antibodies

  • Postpartum thyroiditis:

    • Transient appearance of TPOAbs with fluctuating titers

    • Epitope spreading observed during disease progression

    • Changes in TPOAb characteristics during disease course

When investigating TPO autoantibodies, researchers should carefully characterize the patient population, including disease stage, treatment history, and co-existing autoantibodies. For comparative studies, standardized assays with calibrated reference materials are essential to ensure reproducibility across different cohorts .

What methodologies can differentiate between pathogenic and non-pathogenic TPO antibodies?

Distinguishing between pathogenic and non-pathogenic TPO antibodies requires sophisticated experimental approaches:

  • Functional assays:

    • TPO enzymatic inhibition assays using guaiacol oxidation to measure interference with catalytic activity

    • Complement fixation and activation assays to assess inflammatory potential

    • Antibody-dependent cell-mediated cytotoxicity (ADCC) using thyroid cell cultures

    • Fc receptor binding assays to evaluate effector function potential

  • Epitope specificity analysis:

    • Competition binding experiments with well-characterized monoclonal antibodies

    • Peptide array analysis to map linear epitopes

    • Mutation analysis of key immunodominant regions

    • Structural studies of TPO-antibody complexes using cryo-EM

  • Antibody characteristics assessment:

    • IgG subclass determination (IgG1 and IgG3 have greater pathogenic potential)

    • Affinity measurements using surface plasmon resonance (SPR)

    • Glycosylation profiling of the antibody Fc region

    • Isotype switching analysis in longitudinal patient samples

Research indicates that antibodies targeting specific epitopes within IDR-B may have greater pathogenic potential, particularly those that inhibit TPO enzymatic activity or fix complement. Combining multiple methodological approaches provides the most comprehensive assessment of TPO antibody pathogenicity .

How can molecular modeling approaches advance TPO structure-function research?

Molecular modeling offers powerful tools for TPO research when experimental structural data is limited:

  • Homology modeling techniques:

    • Using related peroxidases (particularly myeloperoxidase) as templates

    • Integration of experimental constraints from antibody binding studies

    • Refinement using molecular dynamics simulations

    • Validation through comparison with low-resolution experimental data

  • Applications of AlphaFold and other AI prediction tools:

    • Generation of domain-specific models with high confidence scores

    • Combining AlphaFold predictions with experimental cryo-EM maps

    • Using AI-predicted structures to guide experimental design

    • Comparative analysis of wild-type and mutant TPO structures

  • Docking approaches for TPO interactions:

    • Substrate and inhibitor docking to predict binding modes

    • Antibody-epitope docking to map recognition interfaces

    • Protein-protein interaction modeling for TPO dimerization

    • Virtual screening for potential therapeutic compounds

  • Model validation strategies:

    • Cross-validation against biochemical data

    • Molecular dynamics stability assessment

    • Epitope accessibility analysis compared with immunological data

    • Energy minimization and structural quality assessment

These computational approaches have successfully predicted the coiled-coil dimerization motif in the C-terminus and helped distinguish between "cis" and "trans" models of TPO orientation. When experimental constraints are incorporated, molecular modeling can generate testable hypotheses about TPO structure-function relationships that guide further research .

What are the latest methodologies for studying TPO in the context of personalized medicine?

Emerging approaches for studying TPO in personalized medicine contexts include:

  • Patient-derived TPO autoantibody repertoire analysis:

    • Single B-cell isolation and antibody cloning from patients

    • Next-generation sequencing of antibody repertoires

    • Phage display libraries to identify patient-specific epitopes

    • Correlation of repertoire features with clinical outcomes

  • Genetic and epigenetic profiling related to TPO:

    • TPO gene polymorphism analysis and correlation with autoimmunity risk

    • Epigenetic modifications affecting TPO expression

    • Integration with genome-wide association studies (GWAS)

    • Transcriptomic analysis of thyroid tissue in different disease states

  • Advanced imaging of TPO in thyroid tissue:

    • Super-resolution microscopy to visualize TPO distribution

    • Multi-color immunofluorescence to assess co-localization with other proteins

    • Tissue mass spectrometry imaging for TPO and hormone distribution

    • Correlative light-electron microscopy for ultrastructural context

  • Therapeutic monitoring approaches:

    • Longitudinal monitoring of TPO antibody characteristics during treatment

    • Epitope-specific antibody profiling to predict treatment response

    • Development of standardized TPO antibody assays with clinical decision thresholds

    • Integration of TPO antibody data with other clinical and laboratory parameters

These methodologies support the development of personalized approaches to thyroid autoimmune disease management, potentially allowing clinicians to predict disease progression, treatment response, and relapse risk based on TPO-related biomarkers .

What are the most promising areas for future TPO human research?

Several promising research directions for human TPO studies include:

  • High-resolution structural determination:

    • Complete cryo-EM structures of full-length TPO in different conformational states

    • Structural characterization of TPO complexed with substrates and inhibitors

    • Comprehensive mapping of conformational epitopes recognized by autoantibodies

    • Structural basis of TPO dimerization and membrane interaction

  • Functional TPO variants in disease:

    • Impact of genetic polymorphisms on TPO structure and function

    • Correlation between TPO variants and autoimmune disease susceptibility

    • Functional consequences of alternative splicing in different tissues

    • Post-translational modification differences in health and disease

  • Novel therapeutic approaches:

    • Development of small molecule modulators of TPO activity

    • Epitope-specific immunotherapy to induce tolerance

    • Prevention of TPO-antibody binding without affecting enzyme function

    • Targeted delivery systems for TPO-directed therapeutics

  • TPO beyond the thyroid:

    • Characterization of TPO expression and function in extra-thyroidal tissues

    • Relationship between TPO autoimmunity and other autoimmune conditions

    • Investigation of TPO's potential roles in cancer and inflammatory diseases

These research areas represent significant opportunities for advancing our understanding of TPO biology and developing novel diagnostic and therapeutic approaches for thyroid disorders and autoimmune diseases .

How can interdisciplinary approaches enhance TPO human research?

Interdisciplinary collaboration offers powerful approaches to advance TPO research:

  • Integration of structural biology with immunology:

    • Combining cryo-EM structures with epitope mapping data

    • Correlating structural features with immunological functions

    • Understanding conformational changes induced by antibody binding

    • Structure-guided design of diagnostic tools and therapeutics

  • Computational biology and artificial intelligence applications:

    • Machine learning for prediction of TPO-antibody interactions

    • Quantum mechanics/molecular mechanics (QM/MM) approaches for enzyme catalysis

    • Network analysis of TPO in thyroid disease pathways

    • AI-assisted design of TPO inhibitors or activity modulators

  • Clinical research integration:

    • Correlation of basic TPO research findings with clinical phenotypes

    • Translational studies of TPO biomarkers in patient cohorts

    • Clinical trials of TPO-targeted therapeutics

    • Patient stratification based on TPO antibody characteristics

  • New technological applications:

    • Single-cell analysis of TPO-reactive B and T cells

    • Organoid models of thyroid tissue for TPO functional studies

    • CRISPR/Cas9 engineering of TPO variants

    • Nanoscale delivery systems for TPO-targeting therapeutics

Product Science Overview

Introduction

Thyroid Peroxidase (TPO) is a crucial enzyme in the thyroid gland, playing a significant role in the synthesis of thyroid hormones. The recombinant form of this enzyme, known as Human Recombinant Thyroid Peroxidase (rTPO), is produced using advanced biotechnological methods to study its properties and applications.

Structure and Classification

Thyroid Peroxidase is a membrane-bound glycoprotein that belongs to the peroxidase family, specifically the XPO subfamily . It contains one EGF-like domain and one Sushi (CCP/SCR) domain . The enzyme is typically found as a homodimer with subunits of approximately 100 kDa molecular weight .

Biological Properties and Functions

TPO is an integral apical membrane glycoprotein of thyroid follicular cells . It is responsible for the iodination of tyrosine residues in thyroglobulin, leading to the production of thyroid hormones such as thyroxine (T4) and triiodothyronine (T3) . These hormones are essential for regulating metabolism, growth, and development.

Recombinant Production

Recombinant Human Thyroid Peroxidase is produced by expressing the extracellular domain of human TPO in insect cells . The recombinant protein is purified to homogeneity using specific monoclonal antibody-based affinity and ion exchange chromatography methods . The recombinant form retains immunochemical properties similar to the native antigen purified from human thyroid glands .

Clinical Significance

Thyroid Peroxidase is one of the main autoantigenic targets in autoimmune thyroid diseases such as Hashimoto’s thyroiditis and Graves’ disease . Anti-TPO autoantibodies are present in 90% of Hashimoto’s thyroiditis and 75% of Graves’ disease patients . Immunoassays for the quantification of these autoantibodies are widely used in clinical practice .

Applications

Recombinant Human Thyroid Peroxidase is utilized as an antigen in assays for the detection of TPO-specific autoantibodies in the blood of patients with autoimmune thyroid diseases . It is also used in research to study the enzyme’s structure, function, and role in thyroid hormone synthesis.

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.