EPO Rat

Erythropoietin Rat Recombinant
Shipped with Ice Packs
In Stock

Description

Production and Formulation

Commercial recombinant rat EPO is available in two primary formats:

Table 1: Formulation Comparison

ParameterWith BSA (1306-RE) Carrier-Free (1306-RE/CF)
Reconstitution10 μg/mL in PBS + 0.1% albumin100 μg/mL in sterile PBS
Stability-20°C; avoid freeze-thaw cycles-20°C; same storage conditions
ApplicationsCell culture, ELISA standardsBSA-sensitive assays

Production systems include:

  • Baculovirus expression: Using stably transfected Sf21 insect cells

  • Mammalian systems: HEK293 cells with C-terminal His-tag purification

Hematopoietic Effects

  • Erythropoiesis: 5,000 U/kg doses increase reticulocytes within 24 hours, peaking at 3 days .

  • Thrombopoiesis: Dose-dependent platelet increases occur via megakaryocyte proliferation (2.4× baseline) .

Neuroprotection

  • Traumatic Brain Injury (TBI):

    • Single 5,000 U/kg IP dose at 24h post-injury reduces hippocampal cell loss by 38% .

    • Triple dosing (Days 1-3) improves spatial learning by 45% vs controls in Morris water maze tests .

Tissue Repair

  • Rotator Cuff Healing: Local EPO injections enhance tendon-bone interface collagen organization, increasing failure load by 22% at 28 days .

Pharmacokinetic Profile

Table 2: Administration Route Comparison

RoutePeak Plasma (Cₘₐₓ)Brain PenetrationTime to Cₘₐₓ
Intraperitoneal10,015 ± 685 mU/mL0.8% of plasma3 hours
Subcutaneous6,200 ± 490 mU/mL0.5% of plasma9 hours

Plasma half-life ranges from 6-9 hours, with detectable brain levels persisting >20 hours post-injection .

Detection and Quantification

The Rat Erythropoietin ELISA Kit (ab274398) demonstrates:

  • Sensitivity: 92-105% recovery across matrices

  • Precision: CV ≤4.2% in cell culture media

  • Dynamic Range: 15.6-1,000 pg/mL

Therapeutic Mechanisms

EPO exerts effects through:

  1. Anti-apoptotic pathways: Blocks caspase-3 activation in erythroid precursors

  2. Angiogenesis induction: Upregulates VEGF in ischemic tissues

  3. Neurogenesis stimulation: Increases doublecortin+ neural progenitors by 3.2× in TBI models

Research Considerations

  • Dose Timing: Delayed administration (24h post-injury) remains effective, challenging previous therapeutic windows .

  • Carrier Proteins: BSA-containing formulations improve stability but may interfere with receptor-binding assays .

  • Species Specificity: Despite 82% human homology, rat EPO shows distinct pharmacokinetics requiring species-matched reagents .

Product Specs

Introduction
Erythropoietin (EPO) is a glycoprotein hormone classified as a type I cytokine. Primarily produced by fibroblast-like cells in the kidney's cortex, EPO regulates red blood cell production by stimulating erythroid differentiation and hemoglobin synthesis. EPO also exhibits neuroprotective properties against brain injuries and anti-apoptotic effects in various tissues.
Description
Recombinant Rat EPO, expressed in HEK293 cells, is a single, glycosylated polypeptide chain consisting of 175 amino acids (27-192 a.a). With a molecular weight of 19.6 kDa, this EPO protein features a 6-amino acid His-tag at the C-terminus and is purified using proprietary chromatographic techniques.
Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation
The EPO protein solution (1mg/ml) is formulated with Phosphate-Buffered Saline at pH 7.4 and contains 10% glycerol.
Stability
For short-term storage (2-4 weeks), keep at 4°C. For extended periods, store frozen at -20°C. Adding a carrier protein (0.1% HSA or BSA) is advisable for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
Purity exceeds 95.0% as determined by SDS-PAGE analysis.
Biological Activity
Biological activity is evaluated through a cell proliferation assay using TF-1 human erythroleukemic cells. The ED50 is within the range of ≤ 2ng/ml.
Synonyms

Erythropoietin-Alpha, EPO-a, EPO-alpha, EP, MGC138142. 

Source

HEK293 cells.

Amino Acid Sequence

DGSAPPRLIC DSRVLERYIL EAKEAENVTM GCAEGPRLSE NITVPDTKVN FYAWKRMKVE EQAVEVWQGL SLLSEAILQA QALQANSSQP PESLQLHIDK AISGLRSLTS LLRVLGAQKE LMSPPDATQA APLRTLTADT FCKLFRVYSN FLRGKLKLYT GEACRRGDRH HHHHH.

Q&A

What are the primary physiological roles of EPO in rats?

EPO functions as a pleiotropic cytokine in rats, similar to humans. While originally identified for its critical role in erythropoiesis (red blood cell production), research has revealed that EPO has significant tissue-protective properties in various organ systems. In rats, EPO is primarily produced in the kidneys in response to hypoxia, with EPO mRNA also expressed in the central nervous system, lungs, and spleen . Its primary role involves regulating erythrocyte production to optimize tissue oxygenation when local O₂ tension drops, which stabilizes hypoxia-inducible factor that binds to hypoxia-responsive elements of the EPO gene to activate transcription . Beyond erythropoiesis, EPO exhibits significant cytoprotective, anti-apoptotic, and anti-inflammatory effects in various rat tissues, particularly in models of ischemia/reperfusion injury, making it valuable for studying potential therapeutic applications.

How do EPO receptors function in rat tissue systems?

EPO requires receptors (EPO-R) to perform its functions in rat tissues. EPO-R is expressed on erythroid cell progenitors and in various tissues including the brain, retina, heart, kidney, vascular smooth muscle cells, myoblasts, and vascular endothelium . Administration of EPO up-regulates EPO-R expression and increases endothelial nitric oxide production.

For tissue-protective effects, accumulating evidence suggests EPO works through a heteroreceptor EPO-R isoform, which comprises a classic EPO-R homodimer and the cytokine β-common receptor (βcR) . This heteroreceptor configuration appears necessary for certain protective effects, as demonstrated in studies of spinal cord injury recovery where both the βcR subunit and EPO-R association were required for motor function recovery . The βcR is also involved in EPO-mediated endothelial nitric oxide synthase (eNOS) activation in endothelial cells , suggesting multiple signaling pathways contribute to EPO's diverse effects in rat tissues.

What are the main differences between endogenous and recombinant EPO in rat studies?

In rat studies, researchers primarily use recombinant human EPO (rhEPO) or plasmid-based EPO gene delivery rather than purified rat EPO. The dosages of rhEPO typically range from 1,000-5,000 IU/kg depending on the experimental model and desired effect. In the carbon monoxide cardiotoxicity study, researchers administered 5,000 IU/kg via intraperitoneal injection , while similar doses were used in the experimental autoimmune neuritis model .

Endogenous EPO production in rats occurs primarily in the kidneys in response to hypoxia, with normal serum levels around 15-20 mU/ml as observed in control rats . When using gene electrotransfer methods, serum EPO levels can increase significantly, with peaks of approximately 68 mU/ml reported after muscle-targeted EPO gene delivery . This represents a substantial increase over baseline levels, enabling researchers to study both physiological and pharmacological effects of EPO elevation.

Methodologically, recombinant EPO administration provides precise dosing control but requires repeated injections, while gene electrotransfer approaches can provide sustained EPO expression over longer periods from a single intervention, making them useful for chronic disease models.

What are the optimal administration protocols for EPO in rat experimental models?

The administration of EPO in rat models varies depending on research objectives, disease model, and desired outcomes. Based on the literature, several effective protocols have been established:

Dosing Regimens:

  • Acute injury models: 5,000 IU/kg administered intraperitoneally as a single dose has shown efficacy in carbon monoxide poisoning models .

  • Autoimmune and inflammatory models: Daily administration of 5,000 IU/kg intraperitoneally, starting either preventively (day 3 post-immunization) or therapeutically (day 10 post-immunization) in experimental autoimmune neuritis .

Administration Routes:

  • Intraperitoneal injection: Most commonly used in rat studies due to ease of administration and good systemic distribution .

  • Gene electrotransfer: A single application of EPO-expressing plasmid (e.g., pCAGGS-Epo) into rat muscle using in vivo electroporation can provide sustained EPO production, with significant elevation of serum EPO levels . This approach is particularly valuable for chronic disease models like renal failure.

Timing is critical - in the carbon monoxide cardiotoxicity model, EPO was administered immediately after CO exposure followed by re-oxygenation with ambient air , while in autoimmune neuritis models, both preventive and therapeutic timing protocols showed efficacy but with different outcomes .

How can researchers effectively measure EPO levels and activity in rat experimental models?

Accurate measurement of EPO levels and assessment of its biological activity in rat models require multiple complementary approaches:

Serum EPO Quantification:

  • ELISA assays: Standard method to measure circulating EPO protein levels in rat serum. In gene electrotransfer studies, baseline serum EPO levels in control rats were approximately 15.2 ± 1.2 mU/ml, while treated rats showed peak levels of 68.0 ± 1 mU/ml .

Tissue EPO and EPO-R Expression:

  • RT-PCR: For quantifying EPO and EPO-R mRNA expression in various tissues.

  • Immunohistochemistry: To visualize EPO and EPO-R protein expression patterns in tissue sections, enabling localization within specific cell types.

  • Western blotting: For semi-quantitative assessment of EPO and EPO-R protein levels in tissue homogenates.

Functional Assessments:

  • Hematological parameters: Including red blood cell count, hemoglobin, and hematocrit to assess erythropoietic effects.

  • Tissue-specific outcomes: Such as myocardial injury assessment via histopathology and electron microscopy as used in carbon monoxide cardiotoxicity studies , or clinical scoring systems for neurological function in experimental autoimmune neuritis .

  • Molecular markers of EPO activity: Including assessment of downstream signaling pathways such as JAK2/STAT5, PI3K/Akt, and MAPK activation.

The combination of these measurements provides comprehensive insight into both the pharmacokinetics and pharmacodynamics of EPO in rat experimental models.

What gene transfer techniques are most effective for studying EPO in rat models?

Gene transfer techniques offer significant advantages for studying EPO in rat models, particularly for chronic conditions requiring sustained EPO expression. Based on the literature, in vivo electroporation of EPO-expressing plasmids into rat muscle has emerged as a particularly effective approach:

Muscle-Targeted Gene Electrotransfer:

  • The pCAGGS-Epo plasmid containing rat EPO cDNA under control of the CAG (cytomegalovirus immediate-early enhancer/chicken β-actin hybrid) promoter has shown reliable expression in rat studies .

  • This approach involves injection of plasmid DNA into muscle tissue followed by application of electrical pulses to enhance cellular uptake and expression.

  • Advantages include long-term production of significant amounts of EPO in circulation from a single application, avoiding the need for repeated injections .

Procedural Considerations:

  • In models of adenine-induced renal failure, muscle-targeted gene transfer with pCAGGS-Epo has successfully increased serum EPO levels and improved anemia associated with chronic renal failure .

  • Similar success has been reported in five-sixths nephrectomy models of renal failure .

  • Both direct injection into surgically exposed muscles and less invasive approaches have been documented.

This gene transfer technique is particularly valuable for studying EPO's effects in chronic disease models, as it provides physiologically relevant expression patterns and sustained therapeutic levels without requiring repeated interventions that may introduce experimental variables.

What are the cardioprotective mechanisms of EPO in rat models of cardiac injury?

EPO demonstrates significant cardioprotective effects in rat models through multiple complementary mechanisms, particularly evident in the carbon monoxide poisoning model:

Structural Protection:

  • EPO administration after CO exposure (5000 IU/kg, intraperitoneal) resulted in significant reduction in cardiomyocyte injury compared to untreated CO-exposed rats (p<0.05) .

  • Histopathological examination revealed that 3000 ppm CO induced extensive myocardium injury with multiple foci of necrosis and lymphocyte infiltration, while EPO treatment significantly reduced these pathological changes .

  • Electron microscopy showed that CO poisoning caused myofibril lysis and mitochondrial swelling in rat myocardium, effects that were attenuated by EPO treatment .

Molecular Mechanisms:
While the exact molecular pathways weren't fully detailed in the carbon monoxide study, other research on EPO's cardioprotective effects in ischemia/reperfusion models suggests EPO works through:

  • Anti-apoptotic signaling: Activation of PI3K/Akt and ERK1/2 pathways

  • Anti-inflammatory effects: Reduction of pro-inflammatory cytokine production

  • Antioxidant effects: Reduction of oxidative stress through enhanced antioxidant enzyme activity

  • Mitochondrial stabilization: Prevention of mitochondrial permeability transition pore opening

These findings indicate EPO has potential as a post-exposure treatment for cardiac injury resulting from carbon monoxide poisoning, acting through multiple cellular protective pathways to preserve cardiomyocyte structure and function .

How does EPO affect neurological outcomes in rat models of brain injury?

EPO exhibits significant neuroprotective properties in rat models of various neurological injuries, working through multiple mechanisms:

Traumatic Brain Injury:

  • In a rat model of pediatric traumatic brain injury using controlled cortical impact, EPO administration improved cognitive outcomes and decreased hippocampal caspase activity .

  • This suggests EPO provides neuroprotection partly through inhibition of apoptotic pathways in the hippocampus, a region critical for learning and memory .

Experimental Autoimmune Neuritis:

  • EPO treatment reduced clinical disease severity in this rat model of human Guillain-Barré syndrome .

  • When administered therapeutically (after disease onset), EPO shortened the recovery phase .

  • Histological examination showed decreased inflammation within peripheral nerves and better maintenance of myelin in EPO-treated animals .

Immunomodulatory Mechanisms:

  • Interestingly, EPO increased the number of macrophages in later stages of experimental autoimmune neuritis .

  • The anti-inflammatory cytokine transforming growth factor (TGF)-beta was upregulated in EPO-treated cohorts .

  • In vitro experiments showed reduced T cell proliferation in the presence of EPO, with moderate induction of TGF-beta .

These findings suggest EPO exerts neuroprotection through both direct cellular protection (anti-apoptotic effects) and immunomodulatory mechanisms, including the induction of beneficial macrophage phenotypes and shifting the immune response toward anti-inflammatory profiles in the peripheral nervous system .

What molecular signaling pathways mediate EPO's tissue-protective effects in rat models?

EPO activates multiple signaling cascades to confer tissue protection in rat models, with pathway engagement varying by tissue type and injury model:

Classical EPO Receptor Signaling:

  • Binding to the homodimeric EPO receptor activates Janus kinase 2 (JAK2), leading to phosphorylation and activation of signal transducer and activator of transcription 5 (STAT5) .

  • This classical pathway primarily mediates erythropoietic effects but also contributes to some tissue-protective functions.

Heteroreceptor Complex Signaling:

  • For many tissue-protective effects, EPO signals through a heteroreceptor complex comprising the EPO receptor homodimer and the β-common receptor (βcR) .

  • In rat models of spinal cord injury, both the EPO-R and βcR components were necessary for recovery of motor function .

  • The βcR is also involved in EPO-mediated endothelial nitric oxide synthase (eNOS) activation in endothelial cells .

Downstream Protective Pathways:

  • Activation of phosphatidylinositol 3-kinase (PI3K)/Akt pathway, which inhibits apoptosis by phosphorylating and inactivating pro-apoptotic proteins.

  • Mitogen-activated protein kinase (MAPK) cascade activation, promoting cell survival and proliferation.

  • Nuclear factor kappa B (NF-κB) modulation, affecting inflammatory responses.

  • In models of autoimmune neuritis, EPO induced anti-inflammatory transforming growth factor (TGF)-beta in macrophages, shifting immune responses toward resolution .

These diverse signaling mechanisms collectively contribute to EPO's cytoprotective, anti-apoptotic, and anti-inflammatory effects observed across various rat tissue injury models, explaining its broad spectrum of tissue-protective properties.

How effective is EPO in rat models of chronic kidney disease and renal anemia?

EPO has demonstrated significant efficacy in addressing both renal dysfunction and associated anemia in rat models of chronic kidney disease:

Gene Electrotransfer Approach:

  • In rats with adenine-induced renal failure, muscle-targeted EPO gene electrotransfer using pCAGGS-Epo plasmid effectively increased serum EPO levels .

  • Similar positive results were observed in five-sixths nephrectomized rat models of chronic renal failure .

  • This gene transfer approach provided sustained EPO expression, avoiding the need for repeated injections.

Outcomes in Renal Anemia:

Translational Considerations:

  • The differential regulation of EPO-R expression has been observed between young, healthy versus older, co-morbid animals, which may partially explain variable responses to EPO therapy .

  • These findings highlight the importance of considering age and comorbidities in experimental designs when studying EPO's effects in renal disease models.

These results suggest that both recombinant EPO administration and gene therapy approaches can effectively address renal anemia in rat models, though response heterogeneity exists and may depend on factors such as age, comorbidities, and specific renal pathology.

What is the efficacy of EPO in rat models of autoimmune neurological disorders?

EPO demonstrates substantial therapeutic potential in rat models of autoimmune neurological disorders, particularly in experimental autoimmune neuritis (EAN), which serves as a model for human Guillain-Barré syndrome:

Clinical Efficacy:

  • In the EAN rat model, EPO treatment reduced clinical disease severity .

  • When administered therapeutically (starting at day 10 after immunization), EPO shortened the recovery phase of the disease .

  • Daily administration of EPO at 5000 IU/kg/day intraperitoneally was effective in both preventive and therapeutic paradigms .

Histopathological Improvements:

  • Clinical improvements were mirrored by decreased inflammation within the peripheral nerves .

  • Myelin was better maintained in EPO-treated animals compared to controls .

Immunomodulatory Mechanisms:

  • Interestingly, EPO increased the number of macrophages, especially in later stages of the experimental disease phase .

  • This finding appears paradoxical but suggests EPO may promote a shift toward beneficial, repair-oriented macrophage phenotypes.

  • The anti-inflammatory cytokine transforming growth factor (TGF)-beta was upregulated in EPO-treated cohorts .

  • In vitro experiments revealed less proliferation of T cells in the presence of EPO, while TGF-beta was moderately induced .

  • Other cytokine secretion was minimally altered by EPO, suggesting specific immunomodulatory effects rather than general immunosuppression .

These findings indicate that EPO exerts its beneficial effects in autoimmune neurological disorders primarily through immunomodulation—specifically by inducing beneficial macrophage phenotypes and shifting the immune system toward anti-inflammatory responses in the peripheral nervous system . This mechanism differs from its classical erythropoietic effects and highlights EPO's pleiotropic nature.

How does EPO administration affect carbon monoxide-induced tissue damage in rat models?

EPO demonstrates significant protective effects against carbon monoxide (CO)-induced tissue damage in rat models, particularly in the myocardium:

Experimental Protocol:

  • Severe carbon monoxide toxicity was induced by exposing Wistar rats to 3000 ppm CO for 40 minutes .

  • EPO (5000 IU/kg) was administered via intraperitoneal injection immediately after CO exposure .

  • Animals were then re-oxygenated with ambient air for recovery .

Cardioprotective Effects:

  • Histopathological examination revealed that 3000 ppm CO induced significant myocardium injury with multiple foci of necrosis and lymphocyte infiltration compared to controls (p<0.05) .

  • Electron microscopy showed myofibril lysis and mitochondrial swelling in the myocardium due to CO poisoning .

  • EPO administration after CO exposure significantly reduced cardiomyocyte injury (p<0.05) .

Mechanistic Insights:

  • The protective effect was observed when EPO was administered post-exposure, suggesting potential therapeutic value even after CO poisoning has occurred .

  • The structural preservation of cardiomyocytes and mitochondria suggests EPO may stabilize cellular and subcellular membranes against CO-induced damage .

  • These findings build upon previous work by the same research group, confirming EPO's protective effect on CO cardiotoxicity .

This research provides compelling evidence that EPO may serve as a therapeutic option for cardiac protection following CO poisoning, with potential clinical applications for treating patients with CO-induced cardiac injury. The effectiveness of post-exposure treatment is particularly promising from a translational perspective .

Why do EPO effects often differ between rat models and human clinical trials?

The discrepancy between promising results in rat models and disappointing outcomes in human clinical trials of EPO can be attributed to several key factors:

Model Limitations:

  • Most preclinical studies use young, healthy animals, while clinical trials enroll patients of variable age with underlying chronic co-morbidities .

  • The controlled laboratory conditions of rat experiments rarely capture the complexity and heterogeneity of human disease states .

  • As noted in the literature, "The following viewpoint uses rhEPO as an example to highlight the possible pitfalls in current practice using young healthy animals for the evaluation of therapies to treat patients of variable age and underlying chronic co-morbidity" .

Species Differences:

  • EPO efficacy appears more pronounced in rodent models than in large animal models, suggesting a species-dependent response gradient .

  • One porcine study even reported complete failure of EPO to exert any cardioprotective effect, highlighting potential species-specific response variations .

  • The literature notes: "It is interesting to note that in the majority of these studies, EPO had more pronounced therapeutic effects in rodents than in large animal models" .

Clinical Complexity:

  • The phenomenon of "EPO resistance" affects approximately 10% of chronic kidney disease patients, characterized by requiring higher doses to maintain recommended hemoglobin levels or lacking response entirely .

  • Differential regulation of EPO-R expression has been observed between young, healthy versus older, co-morbid animals, suggesting age and comorbidity effects on EPO responsiveness .

  • The resuscitative measures and complex clinical management in human patients may modify EPO effects compared to the more straightforward rat models .

These limitations highlight the need for more sophisticated animal models that better recapitulate the complexity of human disease states, particularly incorporating age-related changes and comorbidities when evaluating EPO's therapeutic potential.

What methodological considerations should researchers address when designing EPO rat studies with translational goals?

To maximize translational relevance of EPO studies in rat models, researchers should address several critical methodological considerations:

Model Selection and Refinement:

  • Incorporate age-appropriate rats rather than exclusively using young animals, as EPO response varies with age .

  • Include rats with relevant comorbidities (e.g., diabetes, hypertension) to better mimic human patient populations .

  • Consider using multiple rat strains to account for genetic variability in EPO responsiveness.

Dosing and Administration Protocols:

  • Explore dose-response relationships thoroughly, as the therapeutic window may differ between rats and humans.

  • Test both preventive and therapeutic administration timing to distinguish between mechanisms relevant to prophylaxis versus treatment .

  • Consider clinically relevant administration routes that can be practically implemented in human patients.

Comprehensive Outcome Assessment:

  • Measure both intended therapeutic effects (e.g., tissue protection) and potential adverse effects (e.g., thrombosis, hypertension).

  • Include long-term follow-up to detect delayed benefits or toxicities that might not be apparent in short-term studies.

  • Assess multiple organs systems even when targeting specific tissues, as EPO has pleiotropic effects throughout the body.

Mechanistic Investigations:

  • Examine EPO and EPO-R expression patterns across tissues and compare with human expression profiles.

  • Investigate the heteroreceptor EPO-R/βcR complex that mediates tissue-protective effects versus classical EPO-R homodimers .

  • Characterize downstream signaling pathways activated by EPO in different tissues and disease states.

Reproducibility and Reporting:

  • Implement blinded assessment of outcomes to minimize bias.

  • Pre-register study protocols with clearly defined primary and secondary endpoints.

  • Report negative findings to address publication bias in the literature.

By addressing these methodological considerations, researchers can design rat studies with greater translational potential, potentially bridging the gap between promising preclinical findings and clinical efficacy.

What are the key differences in EPO receptor expression and signaling between healthy and diseased rat models?

EPO receptor expression and signaling pathways exhibit important differences between healthy and diseased rat states, which may explain variable responses to EPO therapy:

EPO-R Expression Patterns:

  • EPO-R expression is dynamically regulated and changes in response to tissue injury or disease states.

  • In renal disease models, differential regulation of EPO-R expression has been observed between young, healthy versus older, co-morbid animals .

  • Children with acute kidney injury presented with elevated EPO-R expression in the kidney despite decreased EPO plasma levels, suggesting compensatory upregulation of receptors during disease .

Heteroreceptor Complex Formation:

  • The tissue-protective effects of EPO appear to be mediated through a heteroreceptor complex comprising the classic EPO-R homodimer and the β-common receptor (βcR) .

  • In injury models, the association between EPO-R and βcR appears critical for functional recovery, as demonstrated in spinal cord injury studies .

  • The relative expression and association of these receptor components may differ between healthy and diseased states, affecting EPO responsiveness.

Signaling Pathway Alterations:

  • Disease states often alter baseline activation of signaling pathways downstream of EPO-R.

  • In inflammatory conditions, baseline activation of JAK/STAT, PI3K/Akt, and MAPK pathways may modify cellular responses to EPO.

  • In the experimental autoimmune neuritis model, EPO induced transforming growth factor (TGF)-beta in macrophages, representing a disease-specific response not typically observed in healthy rats .

Functional Consequences:

  • These receptor and signaling differences may explain observations of "EPO resistance" in certain disease states .

  • They also suggest that EPO doses effective in healthy animals may require adjustment in disease models.

  • The temporal dynamics of EPO-R expression during disease progression may create windows of enhanced or diminished EPO responsiveness.

Product Science Overview

Recombinant Erythropoietin Production

Recombinant erythropoietin (rhEPO) is produced using cells transfected with the EPO gene or EPO cDNA linked to an expression vector . This recombinant DNA is integrated into the genome of the host cell, which then stably expresses the EPO protein over time . The production of rhEPO typically involves mammalian host cells, such as Chinese hamster ovary (CHO) cells, due to the complex glycosylation patterns required for its biological activity .

Glycosylation and Biological Activity

Glycosylation is a critical aspect of EPO’s structure and function. EPO has three N-glycosylation sites, four alpha helices, and an N- to C-terminal disulfide bond that are conserved across species . The glycosylation of EPO is essential for its biological activities in vivo, including its therapeutic efficacy, in vivo activity, and half-life . The glycosylation pattern of glycoproteins is species-dependent, which is why mammalian cells are preferred for producing glycosylated biopharmaceuticals .

Applications of Recombinant Erythropoietin

Recombinant human erythropoietin (rhEPO) is widely used as a therapeutic agent for treating anemia, particularly in patients with chronic kidney disease (CKD), malignancies, and AIDS . It also supports autologous blood collection and represents one of the largest markets for biopharmaceuticals . The administration of rhEPO and its analogues provides significant benefits in preventing and reversing anemia in these conditions .

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.