EPOR Human, Active

Erythropoietin Receptor Human Recombinant, Active
Shipped with Ice Packs
In Stock

Description

Introduction to EPOR Human, Active

EPOR (Erythropoietin Receptor) Human, Active refers to the recombinant, functionally active form of the human erythropoietin receptor. This glycoprotein mediates erythropoietin (EPO)-dependent signaling, critical for erythroid progenitor cell survival, proliferation, and differentiation . Recombinant EPOR is widely used in biochemical and biomedical research to study erythropoiesis, neuroprotection, and metabolic regulation .

Production Methods

EPOR Human, Active is produced in two primary expression systems:

  1. Sf9 Baculovirus Cells:

    • Yields a glycosylated protein (25.6 kDa) in a phosphate-buffered saline (PBS) solution with 10% glycerol .

    • Purity: >95% by SDS-PAGE .

  2. HEK293 Cells:

    • Produces a 26.3 kDa protein (34 kDa under non-reducing conditions) with >98% purity .

    • Lyophilized from sterile PBS (pH 7.4) .

Biological Activity

EPOR binds EPO with high affinity, activating intracellular pathways such as JAK2/STAT, PI3K/Akt, and Ras/MAPK . Key functional data:

  • Activity Assay: Inhibits EPO-dependent proliferation of TF-1 erythroleukemic cells (ED₅₀ = 15–70 ng/mL) .

  • Signaling: Prevents apoptosis in erythroblasts and neurons; enhances synaptic plasticity in hippocampal neurons .

Table 2: Functional Data for EPOR Human, Active

ParameterValueSource
EC₅₀ (TF-1 Inhibition)15–60 ng/mL (HEK293-derived)
Binding Affinity (Kd)~1 nM (high-affinity site)
Endotoxin Levels<1.0 EU/µg (LAL assay)

Research Applications

  • Neuroprotection: Transgenic mice expressing constitutively active EPOR in hippocampal neurons exhibit enhanced spatial learning and long-term potentiation (LTP) .

  • Metabolic Regulation: EPOR in white adipose tissue (WAT) reduces lipogenic gene expression via RUNX1 activation, improving glucose tolerance .

  • Cancer Biology: Dysregulated EPOR signaling is linked to tumor angiogenesis and invasion .

Physical and Chemical Properties

  • Formulation:

    • Liquid: 0.5 mg/mL in PBS (pH 7.4) with 10% glycerol .

    • Lyophilized: Reconstituted in sterile water or PBS .

  • Stability:

    • Liquid: Stable for 2–7 days at 4°C; 3 months at -20°C .

    • Lyophilized: Stable for 12 months at -80°C .

Product Specs

Introduction
The erythropoietin receptor, also known as EPOR, plays a crucial role in the development of red blood cells. It facilitates the proliferation and differentiation of erythroblasts in response to erythropoietin. Upon binding with EPO, EPOR activates the Jak2 tyrosine kinase, which in turn sets off a cascade of intracellular signaling pathways. These pathways include Ras/MAP kinase, phosphatidylinositol 3-kinase, and STAT transcription factors. Activated EPOR also contributes to the survival of erythroid cells. Mutations in the EPOR gene can lead to disorders like erythroleukemia and familial erythrocytosis. Additionally, dysregulation of EPOR can influence the growth of certain tumors.
Description
Produced in Sf9 insect cells using baculovirus expression system, this recombinant EPOR is a single, glycosylated polypeptide chain. It consists of 232 amino acids (25-250a.a.), resulting in a molecular mass of 25.6 kDa. However, on SDS-PAGE, it appears as a band at approximately 28-40 kDa due to glycosylation. This EPOR protein features a 6-amino acid His-tag at its C-terminus, facilitating purification through proprietary chromatographic techniques.
Physical Appearance
A clear, sterile-filtered solution.
Formulation
The EPOR protein is supplied in a solution containing 0.5 mg/ml EPOR, Phosphate Buffered Saline (pH 7.4), and 10% glycerol.
Stability
For short-term storage (up to 2-4 weeks), the EPOR protein should be stored at 4°C. For extended storage, it is recommended to freeze the protein at -20°C. Adding a carrier protein (0.1% HSA or BSA) is advisable for long-term storage. Avoid repeated freeze-thaw cycles to preserve protein integrity.
Purity
The purity of EPOR protein is greater than 95%, as determined by SDS-PAGE analysis.
Biological Activity
The biological activity of EPOR is assessed through its ability to inhibit EPO-dependent proliferation of TF-1 human erythroleukemic cells. The ED50 for this inhibitory effect is less than or equal to 70 ng/ml.
Synonyms
EPO-R, EPOR, Erythropoietin Receptor.
Source
Sf9, Baculovirus cells.
Amino Acid Sequence
APPPNLPDPK FESKAALLAA RGPEELLCFT ERLEDLVCFW EEAASAGVGP GNYSFSYQLE DEPWKLCRLH QAPTARGAVR FWCSLPTADT SSFVPLELRV TAASGAPRYH RVIHINEVVL LDAPVGLVAR LADESGHVVL RWLPPPETPM TSHIRYEVDV SAGNGAGSVQ RVEILEGRTE CVLSNLRGRT RYTFAVRARM AEPSFGGFWS AWSEPVSLLT PSDLDPHHHH HH.

Q&A

What is the molecular structure of human EPOR and how does it function?

Human EPOR is a type I single-transmembrane cytokine receptor belonging to the homodimerizing subclass that functions as ligand-induced or ligand-stabilized homodimers. The receptor has a calculated molecular mass of 59 kDa and consists of:

  • An extracellular domain (ECD) responsible for ligand binding, containing:

    • A single cytokine homology module (CHM) formed by two fibronectin type III (FnIII) domains (D1 and D2)

    • Membrane-distal D1 domain with two conserved disulfides

    • Membrane-proximal D2 domain featuring a characteristic WSXWS motif

  • A single-helical transmembrane domain (TMD) driving receptor dimerization

  • A disordered intracellular domain (ICD) responsible for JAK2 binding and downstream signaling

When EPO binds to EPOR, it induces a conformational change initiating EPOR-associated JAK2 transphosphorylation and activating multiple downstream signal transduction cascades including STATs, PI3K/AKT, RAS/ERK1/2, and NF-κB pathways . These signaling cascades lead to activation of antiapoptotic factors, stimulation of cell differentiation, and modulation of cellular plasticity in a context-dependent manner .

Where is EPOR expressed beyond hematopoietic tissues?

While EPOR was initially characterized in erythroid progenitor cells, research has demonstrated its expression in multiple non-hematopoietic tissues:

  • Central nervous system: neurons, oligodendroglia, astrocytes, and microglia

  • Adipose tissue: both white adipose tissue (WAT) and brown adipose tissue (BAT)

  • Skeletal muscle

  • Cardiovascular system

The widespread expression of EPOR provides the molecular basis for EPO's pleiotropic effects beyond erythropoiesis, including neuroprotection, metabolic regulation, and tissue repair after injury .

How can researchers reliably detect EPOR expression in experimental systems?

Detecting EPOR has been historically challenging due to:

  • Low cell surface expression, even in stimulated states

  • Cross-reactivity issues with commercially available antibodies

  • Scientific disputes regarding expression in extra-hematopoietic tissues

Methodological approaches for reliable EPOR detection:

  • Validated specific antibodies: Use newly developed and thoroughly characterized antibodies that have been validated using EPOR-transduced cells as positive controls

  • Double-labeling strategies: Implement techniques that can demonstrate both membrane expression and intracellular localization (e.g., in the Golgi apparatus)

  • Recombinant protein controls: Utilize high-purity recombinant EPOR proteins (>98% as determined by SDS-PAGE) as positive controls in detection assays

  • Multi-method approach: Combine protein detection (Western blot, immunohistochemistry) with functional assays and gene expression analysis

Implementing these approaches helps overcome the technical challenges that have complicated EPOR research and enables more reliable characterization of EPOR expression patterns.

What is the current understanding of EPOR's role in metabolic regulation?

Recent research has revealed that EPOR plays a significant role in energy metabolism, particularly through its expression in adipose tissue. Key findings include:

  • Adipose tissue-specific functions:

    • Male mice lacking EpoR in adipose tissue exhibit increased fat mass and greater susceptibility to diet-induced obesity

    • EpoR expression in WAT mediates EPO's metabolic activities

  • Metabolic improvements with EPO treatment:

    • Enhanced glucose tolerance and insulin sensitivity

    • Reduced expression of lipogenic-associated genes in WAT

    • Decreased fat mass

  • Molecular mechanism:

    • EPO-EpoR signaling increases transcription factor RUNX1, which directly inhibits lipogenic gene expression

    • EPO treatment decreases WAT ubiquitin ligase FBXW7 expression and increases RUNX1 stability

    • These effects create an EPO-EpoR-RUNX1 regulatory axis controlling energy metabolism

  • Tissue-specific responses:

    • Effects on fat mass and lipogenic gene expression require adipose tissue EpoR expression

    • Mice with EpoR restricted to erythroid tissue (∆EpoR E-mice) become obese, glucose intolerant, and insulin resistant

These findings suggest potential therapeutic applications for EPO or EPO mimetics in metabolic disorders, while also highlighting the importance of considering tissue-specific EPOR functions.

How does the structural dynamics of EPOR-EPO binding influence receptor activation?

The structural dynamics of EPOR-EPO binding and receptor activation involve a complex series of molecular events:

  • Ligand binding and receptor dimerization:

    • A single EPO molecule binds to two EPOR molecules, forming a homodimeric complex

    • This binding follows a general activation mechanism involving ligand binding to a monomeric receptor followed by receptor dimerization

  • Conformational changes:

    • EPO binding induces rotational movement of the receptor transmembrane (TM) α-helices

    • This movement causes proximity, dimerization, and activation of associated JAK2 subunits

  • Structural modeling insights:

    • Three-dimensional models of EPOR complexes with EPO and JAK2 have been generated using AlphaFold Multimer

    • Due to the large size of these complexes (3220-4074 residues), modeling required stepwise assembly from smaller parts

    • Models were validated through comparisons with published experimental data

  • Activation mechanisms:

    • Models support both active and inactive conformational states

    • These models help explain the molecular basis of oncogenic mutations that may involve non-canonical activation routes

Understanding these structural dynamics provides insights for developing targeted therapies that modulate EPOR signaling and may help explain how mutations in EPOR can lead to pathological conditions.

What approaches are available for engineering novel EPOR-binding proteins?

Engineering novel EPOR-binding proteins represents an important frontier in research with potential therapeutic applications. Approaches include:

  • Key residue grafting strategy:

    • Scanning the Protein Data Bank for suitable scaffold proteins that can accept key interaction residues

    • Grafting the key interaction residues of human erythropoietin that bind to EPOR onto unrelated protein scaffolds

    • Making additional mutations to form stable complexes with EPOR

  • Example of successful design:

    • Rat PLCδ1-PH (pleckstrin homology domain of phospholipase C-δ1) was successfully engineered to bind human EPOR

    • A designed triple mutation of PLCδ1-PH (ERPH1) bound EPOR with high affinity (KD of 24 nM and IC50 of 5.7 μM)

    • The wild-type PLCδ1-PH showed no detectable binding under the same assay conditions

  • Validation methods:

    • In vitro binding assays to determine binding affinities

    • Cell-based assays to confirm functional significance

    • Correlation between computational binding affinities and in vitro measurements

  • Activity assessment:

    • Measuring the ability of engineered proteins to inhibit EPO-dependent proliferation

    • The activity of recombinant EPOR can be measured by its ability to inhibit EPO-dependent proliferation of TF-1 human erythroleukemic cells with an ED50 of typically 15-60 ng/mL

This engineering approach demonstrates promising applications in protein engineering targeting protein-protein interfaces and could lead to novel therapeutic agents that modulate EPOR signaling.

How does EPOR expression change under pathological conditions?

EPOR expression is dynamically regulated and changes under various pathological conditions:

  • Injury-induced expression:

    • In mouse models, stereotactically applied stab wounds to the motor cortex lead to distinct EPOR expression by reactive GFAP-expressing cells in the lesion vicinity

    • This suggests EPOR upregulation as part of the tissue response to injury

  • Neurological disorders:

    • In human epilepsy patients, neurons and oligodendrocytes of the hippocampus strongly express EPOR

    • This may represent an adaptive response to neural tissue distress

  • Potential role in carcinogenesis:

    • EPO and EPOR may be involved in carcinogenesis, angiogenesis, and invasion

    • Oncogenic mutations may involve non-canonical activation routes of the EPOR signaling pathway

  • Metabolic disorders:

    • Mice with EpoR restricted to erythroid tissue (lacking EPOR in other tissues) become obese, glucose intolerant, and insulin resistant

    • This suggests that normal EPOR expression in metabolic tissues is protective against metabolic dysfunction

Understanding these pathological changes in EPOR expression provides insights into disease mechanisms and potential therapeutic opportunities for conditions ranging from neurological disorders to metabolic diseases and cancer.

What are the optimal protocols for producing and purifying recombinant human EPOR?

For researchers working with recombinant human EPOR, the following protocols and specifications are important considerations:

  • Expression system selection:

    • HEK293 cells provide a mammalian expression system that ensures proper folding and post-translational modifications

    • The expressed protein typically has a molecular mass of 26.3 kDa, with apparent molecular mass of 34 kDa due to glycosylation

  • Protein construction design:

    • Optimal construct includes the extracellular domain (Met 1-Pro 250) of human EPOR

    • C-terminal polyhistidine tag facilitates purification

    • Proper signal sequence ensures secretion

  • Purification approach:

    • Affinity chromatography using the His-tag

    • Achieve > 98% purity as determined by reducing SDS-PAGE

  • Quality control metrics:

    • Endotoxin levels should be <1.0 EU per μg as determined by the LAL method

    • Activity measurement by inhibition of EPO-dependent proliferation of TF-1 cells

    • Typical ED50: 15-60 ng/mL in the presence of 0.1 U/mL Recombinant Human EPO

  • Storage and handling:

    • Lyophilized from sterile PBS, pH 7.4

    • Lyophilized proteins are stable for up to 12 months when stored at -20 to -80°C

    • Reconstituted protein solution can be stored at 4-8°C for 2-7 days

    • For longer storage, prepare aliquots to avoid freeze-thaw cycles

Product SpecificationsDetails
Molecular Mass26.3 kDa
Apparent Molecular Mass34 kDa
Purity> 98% by SDS-PAGE
Activity ED5015-60 ng/mL
Endotoxin<1.0 EU per μg
Storage Stability12 months at -20 to -80°C (lyophilized)

Following these specifications ensures the production of high-quality recombinant EPOR suitable for both structural and functional studies.

What experimental models are most appropriate for studying tissue-specific EPOR functions?

Selecting appropriate experimental models is crucial for studying tissue-specific EPOR functions:

  • For neurological studies:

    • Mouse models with stereotactically applied stab wounds to study injury-induced EPOR expression

    • Human tissue samples from patients with neurological disorders (e.g., epilepsy) for clinical relevance

    • Primary cultures of neurons, astrocytes, oligodendrocytes, and microglia to study cell-specific responses

  • For metabolic research:

    • Tissue-specific EPOR knockout mice (e.g., adipose-specific EPOR deletion)

    • Diet-induced obesity models to study EPO/EPOR's protective effects

    • Ex vivo adipose tissue cultures to study direct tissue effects

  • For erythropoiesis studies:

    • TF-1 human erythroleukemic cells for proliferation assays

    • Mice with EPOR restricted to erythroid tissue (∆EpoR E-mice) to distinguish erythroid from non-erythroid effects

    • Bone marrow cultures for studying erythroid progenitor responses

  • For structural and binding studies:

    • Recombinant protein systems with purified components

    • Cell lines expressing EPOR for functional binding assays

    • Computational modeling validated with experimental data

  • For signaling pathway analysis:

    • Reporter cell lines for specific pathway activation

    • Phosphoprotein analysis in relevant tissues after EPO stimulation

    • JAK2 inhibitor studies to confirm signaling specificity

How can computational approaches complement experimental studies of EPOR?

Computational approaches provide valuable complementary tools for EPOR research:

  • Structural modeling applications:

    • AlphaFold Multimer has been used to generate three-dimensional models of EPOR complexes with EPO and JAK2

    • These models support understanding of both active and inactive conformations

    • For large complexes (3220-4074 residues), stepwise assembly from smaller parts with validation against experimental data is recommended

  • Membrane dynamics simulations:

    • Models equilibrated in explicit lipids of the plasma membrane provide insights into EPOR behavior in its native environment

    • These simulations help understand the rotational movement of receptor TM α-helices during activation

  • Protein-protein interface design:

    • Computational scanning of the Protein Data Bank identifies suitable scaffold proteins for engineering

    • In silico prediction of binding affinities shows qualitative correlation with experimental measurements

    • This approach has successfully identified proteins that can be engineered to bind EPOR

  • Oncogenic mutation analysis:

    • Computational models help elucidate the molecular basis of oncogenic mutations

    • They provide insights into non-canonical activation routes

  • Integration with experimental data:

    • Models should be selected and validated through comparisons with published experimental data

    • The combination of computational prediction and experimental validation creates a powerful research approach

These computational approaches not only complement traditional experimental methods but also provide insights that might be difficult to obtain through experimental means alone, particularly for understanding dynamic molecular processes and designing novel therapeutics.

What considerations are important when developing therapeutic agents targeting EPOR?

Developing therapeutic agents targeting EPOR requires careful consideration of several factors:

  • Target specificity challenges:

    • EPOR belongs to the cytokine receptor family with structural similarities to other receptors

    • Ensuring specificity for EPOR over related receptors is crucial to avoid off-target effects

    • Engineering approaches that exploit key interaction residues have shown promise in creating specific EPOR-binding proteins

  • Tissue-specific targeting:

    • EPOR is expressed in multiple tissues with different functions

    • Targeting specific tissue pools of EPOR might be necessary to avoid unwanted effects

    • For example, metabolic effects without erythropoietic effects might be desirable in certain conditions

  • Activation mechanism considerations:

    • Understanding the rotational movement of receptor TM α-helices that causes proximity and activation of JAK2

    • Potential to develop agents that modulate specific aspects of EPOR signaling

    • Consideration of both agonistic and antagonistic approaches depending on the therapeutic goal

  • Delivery strategies:

    • Protein-based therapeutics face delivery challenges, especially for CNS targets

    • Small molecule modulators might offer advantages for certain applications

    • Tissue-specific delivery systems may improve therapeutic index

  • Safety monitoring:

    • Effects on erythropoiesis need careful monitoring to avoid polycythemia

    • Potential carcinogenic effects given EPOR's potential role in carcinogenesis, angiogenesis, and invasion

    • Careful assessment of metabolic effects, especially in patients with existing metabolic disorders

  • Functional validation methods:

    • Measuring ability to inhibit EPO-dependent proliferation of model cell lines like TF-1

    • Typical effective dose ranges (ED50 of 15-60 ng/mL) can serve as benchmarks

    • In vivo models should assess both intended and potential off-target effects

These considerations highlight the complexity of developing EPOR-targeted therapeutics and the importance of a comprehensive understanding of EPOR biology across different tissues and disease states.

Product Science Overview

Introduction

The erythropoietin receptor (EPOR) is a protein that plays a crucial role in the regulation of red blood cell production. It is a member of the cytokine receptor family and is primarily expressed on the surface of erythroid progenitor cells in the bone marrow. The receptor is activated by erythropoietin (EPO), a glycoprotein hormone produced mainly by the kidneys in response to hypoxia (low oxygen levels).

Structure and Function

The erythropoietin receptor is a transmembrane protein composed of an extracellular domain, a single transmembrane helix, and an intracellular domain. The extracellular domain is responsible for binding to erythropoietin, while the intracellular domain is involved in signal transduction. Upon binding of erythropoietin to the receptor, a conformational change occurs, leading to the activation of intracellular signaling pathways that promote the survival, proliferation, and differentiation of erythroid progenitor cells into mature red blood cells .

Recombinant Human Erythropoietin Receptor

Recombinant human erythropoietin receptor (rhEPOR) is produced using recombinant DNA technology. This involves the insertion of the human EPOR gene into a suitable expression system, such as mammalian cells, which then produce the receptor protein. The recombinant receptor retains the same structure and biological activity as the naturally occurring receptor, making it a valuable tool for research and therapeutic applications .

Applications
  1. Research: Recombinant human erythropoietin receptor is widely used in research to study the mechanisms of erythropoiesis (red blood cell production) and to investigate the signaling pathways activated by erythropoietin. It is also used to screen for potential therapeutic agents that can modulate the activity of the receptor.

  2. Therapeutics: The recombinant receptor is used in the development of erythropoiesis-stimulating agents (ESAs) for the treatment of anemia, particularly in patients with chronic kidney disease, cancer, or those undergoing chemotherapy. These agents mimic the action of erythropoietin by binding to the erythropoietin receptor and stimulating red blood cell production .

Mechanism of Action

The binding of erythropoietin to its receptor triggers a cascade of intracellular signaling events. The primary signaling pathway involves the activation of the Janus kinase 2 (JAK2) and the subsequent phosphorylation of the signal transducer and activator of transcription 5 (STAT5). This leads to the transcription of genes that are essential for erythroid progenitor cell survival, proliferation, and differentiation .

Clinical Significance

Mutations or dysregulation of the erythropoietin receptor can lead to various hematological disorders. For example, gain-of-function mutations in the EPOR gene can result in primary familial and congenital polycythemia, a condition characterized by an increased red blood cell mass. Conversely, loss-of-function mutations can lead to anemia .

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.