EPOR Human

Erythropoietin Receptor Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to Erythropoietin Receptor (EPOR)

The erythropoietin receptor (EPOR) is a transmembrane protein encoded by the EPOR gene in humans. It belongs to the cytokine receptor family and mediates erythropoietin (EPO)-dependent signaling, primarily regulating red blood cell production (erythropoiesis) by promoting progenitor survival and differentiation . EPOR is expressed in erythroid progenitors and has roles in non-hematopoietic tissues, including the brain and heart .

Genetic Characteristics

  • Chromosomal location: 19p13.3-p13.2 .

  • Transcript: NM_000121.3 (encodes a 232-amino-acid protein) .

  • Domains: Extracellular ligand-binding domain, transmembrane domain, and cytoplasmic tail with tyrosine residues for signaling .

Protein Properties

  • Molecular weight: 25.6 kDa (unglycosylated); migrates at 28–40 kDa due to glycosylation .

  • Structure: Pre-exists as dimers stabilized by transmembrane domain interactions .

Mechanism of Action

EPOR activation involves:

  1. Ligand binding: EPO induces conformational changes in EPOR dimers .

  2. Jak2 kinase activation: Triggers autophosphorylation and downstream signaling (e.g., STAT5, PI3K/AKT, Ras/ERK pathways) .

  3. Cellular effects:

    • Survival: Suppresses apoptosis in erythroid progenitors via Bcl-xL upregulation .

    • Proliferation: Accelerates erythroblast cell cycling in vitro; in vivo roles remain debated .

    • Differentiation: Promotes erythroblast maturation and enucleation .

Paradoxical Effects

  • EPOR signaling increases red blood cell size despite accelerating cell division .

  • Late-stage erythroblasts lose EPOR dependency for survival but require EPO for terminal maturation .

Genetic Mutations

Mutation TypePhenotypeKey Findings
Truncation (e.g., p.Gln82Ter) Familial erythrocytosis (ECYT1)Impaired negative regulation → constitutive signaling → elevated RBC production
Tyrosine 343 mutations ErythroleukemiaDisrupted STAT5 docking → aberrant proliferation

Mouse Models

  • EPOR knockout: Embryonic lethality (E13.5) due to anemia; rescued by human EPOR transgenes .

  • Truncated EPOR: Mice exhibit polycythemia or anemia, depending on residual tyrosine activity .

Key Studies

  1. Human EPOR in Mice

    • Human EPOR partially rescues erythropoiesis in EPOR-deficient mice but with reduced hematopoietic efficiency .

    • Brain-specific EPOR expression in embryonic mice suggests developmental roles beyond erythropoiesis .

  2. Cell-Cycle Dynamics

    • EPOR shortens erythroblast cell cycles (≤10 hours) in vivo, enhancing erythropoietic output during stress .

  3. Mutation Analysis

    • Truncating mutations (e.g., p.Gln82Ter) elevate serum EPO levels without affecting hemoglobin, implying compensatory mechanisms .

Therapeutic and Diagnostic Relevance

  • Recombinant EPOR: Used to study signaling mechanisms (e.g., Sf9 baculovirus-produced EPOR with His-tag) .

  • Targeted therapies: Jak2 inhibitors (e.g., for polycythemia vera) and EPO mimetics under investigation .

  • Biomarker potential: Elevated EPO/EPOR activity correlates with tumor progression and inflammatory diseases .

Product Specs

Introduction
The erythropoietin receptor, also known as EPOR, plays a crucial role in the production of red blood cells. It facilitates the proliferation and differentiation of erythroblasts upon binding with erythropoietin (EPO). This binding triggers the activation of Jak2 tyrosine kinase, which in turn sets off several intracellular signaling pathways such as Ras/MAP kinase, phosphatidylinositol 3-kinase, and STAT transcription factors. These pathways are essential for erythroid cell survival and development. Mutations in the EPOR gene can lead to blood disorders like erythroleukemia and familial erythrocytosis. Moreover, dysregulation of EPOR can impact the growth of certain tumors.
Description
Recombinant human EPOR, produced in Sf9 insect cells using a baculovirus expression system, is a single glycosylated polypeptide chain. This protein consists of 232 amino acids (25-250 a.a), has a molecular weight of 25.6 kDa, and appears as a band between 28-40 kDa on SDS-PAGE under reducing conditions. This discrepancy in observed versus calculated molecular weight is attributed to glycosylation. The EPOR protein is engineered with a 6-amino acid His-tag at the C-terminus to facilitate purification using chromatographic techniques.
Physical Appearance
The purified EPOR protein solution is clear and sterile-filtered.
Formulation
The EPOR protein is supplied in a solution with a concentration of 0.5 mg/ml. The storage buffer consists of Phosphate Buffered Saline (pH 7.4) and 10% glycerol.
Stability
For short-term storage (up to 2-4 weeks), the EPOR protein solution can be stored at 4°C. For long-term storage, it is recommended to store the protein at -20°C. Adding a carrier protein like HSA or BSA (0.1%) can enhance protein stability during long-term storage. To maintain the integrity of the protein, avoid repeated freeze-thaw cycles.
Purity
The purity of the EPOR protein is greater than 95%, as determined by SDS-PAGE analysis.
Synonyms

Erythropoietin Receptor, EPO-R, EPOR.

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 mediate signaling?

Human Erythropoietin Receptor (EPOR) is a class I cytokine receptor that mediates the effects of erythropoietin (Epo) through a unique dimerization mechanism. The crystal structure, determined at 1.9 Å resolution, reveals that Epo binding imposes a specific 120° angular relationship between two EPOR molecules, which is critical for optimal signaling . This orientation initiates intracellular phosphorylation cascades.

Methodologically, researchers studying EPOR structure should note that signaling efficiency is remarkably sensitive to receptor orientation. The half-maximal response in cellular proliferation assays occurs at an Epo concentration of just 10 pM, which is 10^-2 of its Kd value for the high-affinity binding site (approximately 1 nM) and 10^-5 of the Kd for the low-affinity binding site (approximately 1 μM) . This indicates that only about 6% of cell-surface receptors need to be engaged to achieve half-maximal cellular response, highlighting the efficiency of the EPOR signaling system.

How is EPOR expression regulated in different human tissues?

EPOR expression extends beyond hematopoietic tissues, with significant expression in neural tissues, which explains its roles in neuroprotection . While primarily associated with erythroid precursor cells, EPOR is present in various cell types including neurons and certain cancer cells .

To study tissue-specific EPOR expression, researchers commonly employ:

  • RNA-seq analysis from TCGA databases for comparative expression studies

  • Immunohistochemistry with validated anti-EPOR antibodies

  • Single-cell RNA sequencing to identify cell-specific expression patterns

  • RT-qPCR for quantitative expression analysis in different tissues

Researchers should be aware that antibody specificity for EPOR has been problematic in historical studies, necessitating rigorous validation of reagents before expression analysis.

What are the downstream signaling pathways activated by EPOR?

EPOR activation triggers multiple intracellular signaling cascades primarily through the JAK2-STAT5 pathway. Upon ligand binding and receptor dimerization, the following methodological process occurs:

  • Conformational changes in the receptor dimer with the specific 120° angle positioning

  • JAK2 autophosphorylation

  • Phosphorylation of tyrosine residues on the intracellular domain of EPOR

  • Recruitment and activation of STAT5 and other signaling molecules

  • Activation of secondary pathways including PI3K/AKT and MAPK/ERK

The C-terminal region of EPOR contains negative regulatory domains that recruit phosphatases to terminate signaling. Truncation mutations affecting this region result in prolonged signaling and can lead to primary erythrocytosis .

How does EPOR signaling simultaneously affect cell cycling and red blood cell size?

Recent research has uncovered a paradoxical effect of EPOR signaling on erythropoiesis. Contrary to traditional understanding of cell cycle and size regulation, EPOR signaling increases red cell size while simultaneously increasing both the number and speed of erythroblast cell cycles .

Methodologically, this phenomenon can be studied using:

  • Flow cytometry to measure cell size and cycle progression simultaneously

  • EdU incorporation assays to quantify cell cycle kinetics

  • Mean corpuscular volume (MCV) measurements in animal models with modified EPOR signaling

  • In vivo studies tracking red cell parameters following Epo administration

Research has demonstrated that the increase in MCV persists beyond the duration of Epo treatment and is not simply the result of increased reticulocyte numbers . Importantly, this EPOR-mediated size regulation mechanism operates independently of the established iron-dependent pathways that regulate red cell size.

What are the neuroprotective mechanisms of EPOR signaling in human neurons?

EPOR mediates neuroprotection through several mechanisms that researchers can investigate using specific methodological approaches:

  • Anti-apoptotic effects: EPOR activation in neurons inhibits apoptotic pathways through upregulation of anti-apoptotic proteins

  • Enhanced cognitive performance: Epo has been shown to improve memory functions in both healthy humans and patients with schizophrenia and mood disorders

  • Neuroprotection after hypoxic/ischemic insults: Epo promotes neuronal survival after oxygen deprivation

  • Regenerative effects: EPOR signaling promotes regeneration after axonal damage

Research methodologies to study these mechanisms include:

  • Human induced pluripotent stem cell-derived neurons for survival assays

  • Hypoxia chamber experiments to simulate ischemic conditions

  • Neurobehavioral testing in animal models with modified EPOR expression

  • Clinical studies measuring cognitive outcomes after Epo administration

Recent discoveries indicate that CRLF3 (Cytokine Receptor-Like Factor 3) functions as a novel receptor for Epo variant 3 (EV-3) and plays a crucial role in Epo-mediated neuroprotection independent of classical EPOR signaling .

How do truncated EPOR variants impact erythropoiesis and disease phenotypes?

Truncated EPOR variants have significant implications for erythropoiesis and are associated with primary erythrocytosis. A specific G to A transition in nucleotide 6002 of the EPOR gene causes a TGG (tryptophan) to TAG (stop) codon conversion, resulting in truncation of the 70 C-terminal amino acids of the EPOR molecule .

This truncation affects the receptor by:

  • Removing negative regulatory domains that normally facilitate signal termination

  • Reducing receptor phosphorylation at inhibitory sites

  • Prolonging JAK2-STAT5 signaling

  • Enhancing erythroid precursor sensitivity to Epo

The phenotypic consequence is a mild autosomal dominant form of erythrocytosis. This mutation has been identified in heterozygous form in a large kindred where it cosegregates with the disease phenotype in all 29 affected family members studied .

Research approaches to studying truncated EPOR variants include:

  • CRISPR/Cas9 gene editing to recapitulate specific mutations

  • Phosphoproteomic analysis of signaling duration and intensity

  • In vitro colony-forming assays to measure erythroid progenitor sensitivity to Epo

  • Mouse models expressing human EPOR variants

What is the relationship between EPOR expression and cancer outcomes in pan-cancer analysis?

EPOR expression has emerged as a potential prognostic biomarker across multiple cancer types. Pan-cancer analysis methodologies reveal correlations between EPOR expression and patient survival metrics .

Researchers studying EPOR in cancer contexts should employ:

  • TCGA database analysis for expression and survival correlations

  • Univariate and multivariate Cox regression for survival analysis

  • Kaplan-Meier survival curves stratified by EPOR expression levels

  • Analysis of EPOR expression in relation to clinical parameters

How does EPOR expression correlate with tumor immune microenvironment?

EPOR plays potential roles in tumor immunity through interactions with various immune cell populations. Methodologically, researchers can investigate these relationships by:

  • Analyzing correlations between EPOR expression and six types of tumor-infiltrating immune cells: B cells, CD4+ T cells, CD8+ T cells, macrophages, neutrophils, and dendritic cells

  • Applying algorithms such as CIBERSORT, EPIC, quanTIseq, xCell, and MCP-counter to deconvolute immune cell populations from bulk tumor RNA-seq data

  • Evaluating tumor microenvironment parameters through immune score, stromal score, and estimate score analyses

  • Correlating EPOR expression with tumor mutation burden (TMB) and microsatellite instability (MSI)

These approaches enable researchers to understand how EPOR signaling may modulate anti-tumor immunity and potentially influence immunotherapy responses.

What protein-protein interactions are critical for EPOR function in different cellular contexts?

EPOR engages in numerous protein-protein interactions that influence its signaling capabilities and cellular functions. Key methodological approaches to study these interactions include:

  • Protein-protein interaction (PPI) network analysis using databases like STRING

  • Co-immunoprecipitation followed by mass spectrometry

  • Proximity ligation assays in relevant cell types

  • FRET/BRET analyses for real-time interaction monitoring

Research has identified interactions between EPOR and proteins involved in JAK-STAT signaling, cytoskeletal regulation, and receptor trafficking . Additionally, EPOR forms heteromeric complexes with other receptors like the β common receptor (CD131) in certain tissues, particularly in the nervous system .

How does EPOR splice variant EV-3 signaling differ from classical EPO signaling?

The Epo splice variant EV-3, characterized by the lack of exon 3, mediates neuroprotection through mechanisms distinct from classical EPOR signaling. Unlike full-length Epo, EV-3 appears to act independently of both homodimeric EPOR and heteromeric EPOR/β common receptor complexes .

Methodological approaches to distinguish EV-3 signaling include:

  • Receptor binding assays comparing EV-3 and full-length Epo

  • Signaling studies in cells expressing specific receptor subtypes

  • Neuroprotection assays with receptor-selective antagonists

  • Cross-species conservation analysis of EV-3 responsive elements

Recent research has identified CRLF3 as a receptor for EV-3 that mediates neuroprotection, representing the first known receptor for this Epo variant . This discovery opens new research avenues for developing selective neuroprotective therapies that avoid the erythropoietic side effects of full Epo.

What experimental models best represent human EPOR signaling dynamics?

Researchers investigating human EPOR function must select appropriate experimental models. The following methodological considerations are important:

Model SystemAdvantagesLimitationsBest Applications
Human iPSC-derived erythroid cellsAuthentic human receptor dynamicsVariability between linesErythropoiesis studies
Human iPSC-derived neuronsHuman neuronal EPOR signalingTechnical complexityNeuroprotection research
CRISPR-edited cell linesPrecise genetic manipulationMay lack physiological contextMechanism studies
Humanized mouse modelsIn vivo context with human EPORSpecies differences in signalingTranslational research
Patient-derived samplesDirect clinical relevanceLimited availabilityValidation studies

When designing experiments, researchers should consider that EPOR signaling is highly sensitive to receptor orientation and density. The crystal structure of the Epo-EPOR complex demonstrates that a specific 120° angular relationship is critical for optimal signaling , which may not be perfectly recapitulated in all model systems.

How can researchers distinguish between different EPOR-mediated signaling pathways?

Multiple signaling pathways downstream of EPOR contribute to its diverse biological effects. Methodological approaches to dissect these pathways include:

  • Phospho-specific antibodies and Western blotting to track activation of specific pathways

  • Small molecule inhibitors targeting individual pathway components

  • CRISPR/Cas9 knockout of pathway-specific mediators

  • Phosphoproteomic analysis to identify novel phosphorylation targets

  • Transcriptomic profiling to identify pathway-specific gene expression signatures

Advanced techniques such as single-cell signaling analysis and live-cell biosensors can provide temporal resolution of EPOR signaling dynamics, revealing how different pathways are activated with distinct kinetics following receptor stimulation.

What strategies can minimize erythropoietic effects while preserving neuroprotective benefits of EPOR signaling?

A significant challenge in therapeutic applications of Epo is achieving neuroprotection without unwanted erythropoietic effects. Research methodologies addressing this challenge include:

  • Development of Epo derivatives and mimetics that selectively activate neuroprotective pathways

  • Targeting of alternative receptors like CRLF3 that mediate neuroprotection independently of classical EPOR

  • Structure-based drug design guided by the crystal structure of Epo-EPOR complexes

  • Tissue-specific delivery systems to target neural tissues while minimizing systemic exposure

Studies have demonstrated that some Epo mimetics with partial sequence similarity or unrelated structure compared to full Epo can provide neuroprotective effects without activating homodimeric EPOR . Additionally, naturally occurring Epo variant EV-3 mediates neuroprotection through CRLF3 rather than classical EPOR signaling .

How do genetic variations in EPOR affect patient responses to erythropoiesis-stimulating agents?

Genetic variations in EPOR can significantly impact patient responses to erythropoiesis-stimulating agents (ESAs). Research methodologies to investigate these relationships include:

  • Pharmacogenomic studies correlating EPOR variants with clinical responses

  • Ex vivo erythroid colony assays with patient-derived progenitors

  • Receptor binding and signaling studies using cells expressing variant EPOR

  • Genome-wide association studies to identify additional genetic modifiers

The truncated EPOR variant described in search result causes enhanced sensitivity to Epo, resulting in erythrocytosis. Similar variants may impact patient responses to therapeutic ESAs, potentially requiring dose adjustments to prevent excessive erythropoiesis.

[THIS IS TABLE: Comparison of EPOR Signaling in Different Tissue Contexts]

Tissue ContextPrimary Signaling PathwaysBiological OutcomesKey Research Methods
Erythroid precursorsJAK2-STAT5, PI3K/AKTProliferation, differentiation, survivalCFU-E assays, flow cytometry
NeuronsJAK2-STAT5, PI3K/AKT, NF-κBNeuroprotection, regenerationSurvival assays, neurite outgrowth
Cancer cellsJAK2-STAT5, MAPK/ERKVariable (context-dependent)Proliferation assays, tumor models
Endothelial cellsJAK2-STAT5, PI3K/AKTAngiogenesis, vascular protectionTube formation, migration assays

Product Science Overview

Introduction

The Erythropoietin Receptor (EpoR) is a crucial component in the regulation of red blood cell production. It is a protein found on the surface of erythroid progenitor cells in the bone marrow. The receptor is activated by erythropoietin (EPO), a hormone primarily produced by the kidneys in response to hypoxia (low oxygen levels). The interaction between EPO and EpoR is essential for the survival, proliferation, and differentiation of erythroid progenitor cells into mature red blood cells.

Structure and Function

EpoR is a member of the cytokine receptor family and is composed of a single polypeptide chain. The receptor has an extracellular domain that binds to EPO, a transmembrane domain, and an intracellular domain that transduces the signal into the cell. Upon binding of EPO to EpoR, the receptor undergoes a conformational change that activates intracellular signaling pathways, including the JAK2/STAT5, PI3K/Akt, and MAPK pathways. These pathways lead to the transcription of genes involved in erythroid cell survival and proliferation .

Recombinant Erythropoietin Receptor

Recombinant human erythropoietin receptor (rhEpoR) is produced using recombinant DNA technology. This involves inserting the gene encoding EpoR into a suitable host cell, such as Chinese hamster ovary (CHO) cells, which then express the receptor. The recombinant receptor is used in various research and clinical applications to study the mechanisms of erythropoiesis and to develop therapeutic agents for anemia and other related conditions .

Clinical Applications

The recombinant EpoR is used in the development of erythropoiesis-stimulating agents (ESAs), which are used to treat anemia, particularly in patients with chronic kidney disease, cancer, and those undergoing chemotherapy. ESAs mimic the action of natural EPO by binding to EpoR and stimulating red blood cell production. This reduces the need for blood transfusions and improves the quality of life for patients with anemia .

Research and Development

Research on EpoR has led to significant advancements in understanding the molecular mechanisms of erythropoiesis. Studies have shown that EpoR is not only expressed in erythroid progenitor cells but also in other tissues, including the brain, heart, and kidneys. This suggests that EpoR may have additional roles beyond erythropoiesis, such as neuroprotection and tissue repair .

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.