EPO, Clone PAT1C12AT

Erythropoietin clone PAT1C12AT, Mouse Anti Human
Shipped with Ice Packs
In Stock

Product Specs

Introduction

Erythropoietin (EPO) belongs to the EPO/TPO family of cytokines. This secreted, glycosylated protein consists of 4 alpha helical bundles. EPO, present in plasma, plays a crucial role in red blood cell production. It does so by promoting the differentiation of erythroid cells and initiating the synthesis of hemoglobin. Moreover, EPO exhibits neuroprotective effects against various brain injuries and possesses anti-apoptotic properties in multiple tissue types.

Physical Appearance
The product appears as a colorless solution that has been sterilized through filtration.
Formulation

The product is provided at a concentration of 1mg/ml and is dissolved in a solution composed of PBS with a pH of 7.4, 10% Glycerol, and 0.02% Sodium Azide.

Stability

For storage durations of up to 1 month, maintain the product at a temperature of 4°C. If longer storage is required, store the product at -20°C. It is crucial to avoid subjecting the product to repeated cycles of freezing and thawing.

Synonyms
EPO-a, EPO-alpha, Epoetin, EP, MGC138142.
Purification Method

EPO antibody was purified from mouse ascitic fluids by protein-A affinity chromatography.

Type
Mouse Anti Human Monoclonal.
Clone

PAT1C12AT.

Ig Subclass

Mouse IgG1 heavy chain and k light chain.

Q&A

What is the basic molecular structure and function of erythropoietin (EPO)?

Erythropoietin is a secreted, glycosylated cytokine composed of four alpha helical bundles. It is found in plasma and functions primarily to regulate red cell production through two key mechanisms: promoting erythroid differentiation and initiating hemoglobin synthesis. Beyond its hematopoietic functions, EPO demonstrates neuroprotective activity against various potential brain injuries and exhibits antiapoptotic functions in several tissue types .

The protein's structure is critical to its function, as its four-bundle configuration enables specific binding to its receptor, triggering the signaling cascade necessary for erythropoiesis. This structure-function relationship explains why properly folded recombinant EPO maintains biological activity while denatured forms typically do not.

How is the human erythropoietin receptor gene structured?

The human erythropoietin receptor gene spans approximately 6.5 kilobases with its coding region subdivided by seven intervening sequences (introns) ranging from 81 bp to 2.1 kb in length. The gene's 5' regulatory region contains several notable features:

  • A 123-purine stretch located between positions -456 and -578 upstream from the first codon

  • A palindromic sequence consisting of an almost perfect inverted repeat (CAGCTGC(G/C)TCCG) centered at position -92

  • An inverted SP1 binding site (CCGCCC) at position -151

  • An inverted GATA-1 binding site (TTATCT) at position -179

  • CACCC sequences at position -585 and further upstream

What are the optimal techniques for cloning and expressing functional human EPO?

For successful cloning and expression of functional human EPO, researchers should implement a multi-step methodology:

  • Gene Isolation: Use PCR amplification from human tissue (preferably kidney or liver) with primers designed to capture the complete coding sequence.

  • Vector Selection: Choose mammalian expression vectors containing strong promoters (CMV or EF1α) for optimal expression. Bacterial expression systems often produce non-glycosylated EPO with reduced biological activity.

  • Transfection Optimization: For transient expression, lipofection in HEK293 cells typically yields higher protein levels than electroporation. For stable expression, select CHO cells for their robust protein production capabilities and appropriate post-translational modifications.

  • Purification Strategy: Implement a two-step chromatography approach:

    • Initial capture using immobilized metal affinity chromatography (if tagged)

    • Followed by size exclusion chromatography to achieve >95% purity

  • Activity Verification: Confirm biological activity through in vitro erythroid progenitor cell proliferation assays rather than relying solely on immunological detection methods.

The transcription initiation site appears to be located approximately 132 ± 5 nucleotides downstream from the inverted SP1 site, based on in vitro transcription assays and T1 analysis of human EPO receptor mRNA . This knowledge is crucial for designing expression constructs that maintain natural regulatory elements.

How can researchers troubleshoot EPO expression systems that yield low protein production?

When confronting low EPO expression levels, researchers should systematically evaluate:

  • Plasmid Integrity: Verify complete, error-free coding sequence through sequencing.

  • Codon Optimization: Analyze and optimize codon usage for the host expression system, particularly focusing on rare codons that might limit translation efficiency.

  • Signal Peptide Function: Ensure proper secretion by confirming signal peptide functionality; consider testing alternative signal sequences.

  • Post-translational Modifications: Assess glycosylation patterns using glycosidase treatments followed by western blotting to identify potential processing bottlenecks.

  • Expression Conditions: Optimize temperature, timing, and media formulations:

ParameterStandard ConditionOptimization RangeEffect on EPO Expression
Temperature37°C30-34°CLower temperatures often increase properly folded protein
Harvest Time72 hours48-120 hoursExtended time may increase yield but risk degradation
Media SupplementsStandardAdd 5-10% FBS, 1% ITSCan enhance expression and proper folding
Transfection Ratio1:3 (DNA:reagent)1:2 to 1:5Optimize ratio for each cell line
  • Translational Efficiency: Investigate potential translational repression mechanisms, as studies on other proteins have shown that factors like Paip2 can inhibit translation through interactions with poly(A) binding protein .

What are the essential controls needed for EPO cloning experiments?

A methodologically sound EPO cloning experiment must incorporate these controls:

  • Positive Expression Control: Include a well-characterized gene (e.g., GFP) in a parallel transfection to verify transfection efficiency and expression system functionality.

  • Empty Vector Control: Transfect cells with the expression vector lacking the EPO insert to identify any background effects.

  • Commercial EPO Standard: Use pharmaceutical-grade EPO as a positive control for activity assays and structural analyses.

  • Glycosylation Controls: Include enzymatic deglycosylation treatments to assess the contribution of glycosylation to activity.

  • Antibody Validation: When using anti-EPO antibodies for detection, verify specificity using epitope-mapped monoclonal antibodies like Epo2 clone .

These controls facilitate interpretation of experimental outcomes and help distinguish true biological effects from technical artifacts. Without these controls, researchers risk obtaining misleading results that cannot be properly interpreted within the broader scientific context.

How should researchers design comparative tests to demonstrate EPO variant efficacy?

When evaluating EPO variants, design comparative tests following these methodological principles:

  • Structural Approximation: Ensure the closest possible structural approximation between the variant and reference standards in comparable applications.

  • Distinguishing Feature Isolation: Modify comparison elements so they differ only by the distinguishing feature under investigation, allowing clear attribution of any observed effects to that specific feature .

  • Reproducibility Requirements: Ensure all comparative tests are reproducible based on the provided experimental information.

  • Protocol Standardization: Maintain identical testing conditions for all variants, including:

    • Cell passage number and density

    • Identical media composition and incubation times

    • Consistent protein quantification methods

    • Standardized activity assay parameters

  • Statistical Analysis: Implement appropriate statistical methods for comparing variant performance, typically including:

    • Power analysis to determine sample size

    • Multiple biological replicates (minimum n=3)

    • Appropriate statistical tests (ANOVA with post-hoc analysis)

    • Calculation of effect sizes, not just p-values

This methodological approach follows established case law principles for demonstrating inventive step through comparative testing, where an unexpected effect demonstrated in properly designed comparative tests can provide strong evidence of novelty .

How can researchers distinguish between direct EPO effects and secondary signaling outcomes?

To differentiate primary EPO effects from secondary signaling cascades:

  • Temporal Analysis: Implement time-course experiments capturing very early signaling events (0-30 minutes) compared to later outcomes (hours to days).

  • Pathway Inhibition Strategy: Selectively block downstream signaling components using:

    • JAK2 inhibitors (e.g., AG490) to block the primary signaling kinase

    • STAT5 inhibitors to interrupt the main transcription factor activation

    • PI3K/AKT inhibitors to block the survival/proliferation pathway

  • Receptor Mutagenesis: Generate EPO receptor variants with mutations in specific signaling motifs to dissect pathway-specific contributions.

  • Transcriptome Analysis: Compare immediate-early gene activation (0-2 hours) with secondary transcriptional programs (6-48 hours) using RNA-seq or microarray approaches.

  • Proteomic Identification: Use phosphoproteomics to map the sequence of protein phosphorylation events following EPO receptor activation.

By implementing this multi-layered approach, researchers can construct detailed signaling maps that separate primary receptor-proximal events from secondary and tertiary signaling outcomes, enabling more precise targeting of specific pathway components for therapeutic intervention.

What are the key considerations when establishing an EPO-expressing cell line for long-term research?

When generating stable EPO-expressing cell lines for sustained research:

  • Genomic Integration Strategy:

    • Consider site-specific integration systems (CRISPR/Cas9) rather than random integration

    • Evaluate multiple integration sites to identify optimal expression locations

    • Characterize integration sites to avoid disrupting essential genes

  • Selection System Design:

    • Implement dual selection systems (antibiotic + fluorescent marker)

    • Use antibiotic concentration gradients to identify optimal selection pressure

    • Establish kill curves for the specific cell line being used

  • Expression Stability Assessment:

    • Monitor expression levels over extended passages (minimum 20 passages)

    • Quantify both mRNA and protein levels to identify transcriptional vs post-transcriptional regulation

    • Implement inducible expression systems for toxic or growth-inhibitory constructs

  • Clonality Verification:

    • Perform limiting dilution or FACS-based single-cell sorting

    • Verify monoclonality through genomic PCR and Southern blotting

    • Characterize at least 3-5 independent clones to account for positional effects

  • Functional Validation:

    • Confirm biological activity retention through bioassays at multiple passages

    • Verify protein modifications (glycosylation patterns) remain consistent

    • Establish master and working cell banks with detailed characterization

This methodological approach ensures the development of research tools that provide consistent, reliable EPO expression for extended experimental timeframes, avoiding the common pitfall of expression instability that can compromise long-term studies.

What ethical safeguards should be implemented when working with EPO clones?

Researchers working with EPO clones should implement these ethical safeguards:

  • Dual-Use Awareness: EPO has legitimate research applications but also potential for misuse in sports doping. Researchers should:

    • Implement robust material tracking systems

    • Restrict access to EPO-producing clones

    • Document all distribution of materials or cells

  • Clone Containment: Establish protocols to prevent:

    • Unintended release of genetically modified organisms

    • Cross-contamination between cell lines

    • Accidental human exposure to experimental constructs

  • Research Purpose Limitations: Clearly define and document the scientific purpose of all EPO clone generation, avoiding:

    • Any work aimed at human performance enhancement

    • Development of methods to evade detection in doping tests

    • Creation of clones specifically designed to circumvent regulatory oversight

  • Ethical Review: Submit research proposals to institutional ethics committees, particularly when:

    • Working with primary human tissues

    • Developing novel EPO variants with unknown safety profiles

    • Conducting animal studies involving EPO administration

  • Patent Considerations: Be aware of existing patents on EPO technologies, as patent offices like the European Patent Office (EPO) have specific regulations regarding biological materials .

Implementing these safeguards ensures research integrity while preventing misuse of potentially sensitive biotechnology. This approach aligns with international scientific standards for responsible research conduct.

Product Science Overview

Erythropoietin Clone PAT1C12AT

Clone PAT1C12AT is a monoclonal antibody specifically designed to target human erythropoietin. This clone is derived from the hybridization of mouse F0 myeloma cells with spleen cells from BALB/c mice immunized with human EPO . The antibody is of the IgG1 subclass, with a heavy chain and kappa light chain .

Production and Purification

The production of the EPO antibody involves the hybridoma technique, where the mouse myeloma cells are fused with the spleen cells of immunized mice. This results in the creation of hybrid cells that can produce the desired antibody. The antibody is then purified from mouse ascitic fluids using protein-A affinity chromatography .

Formulation and Stability

The EPO antibody is supplied as a sterile filtered colorless solution, typically containing phosphate-buffered saline (PBS) with a pH of 7.4, 10% glycerol, and 0.02% sodium azide . For optimal stability, the antibody should be stored at 4°C for short-term use (up to 1 month) and at -20°C for long-term storage to prevent freeze-thaw cycles .

Applications

The EPO antibody clone PAT1C12AT is widely used in various research applications, including:

  • Western Blotting: For detecting EPO in different samples.
  • Immunoaffinity Purification: For purifying EPO from complex mixtures.
  • Immunohistochemistry: For studying the localization of EPO in tissues.
Significance

The EPO antibody clone PAT1C12AT is significant in both clinical and research settings. It helps in understanding the role of EPO in erythropoiesis and its potential therapeutic applications. Additionally, EPO has neuroprotective activity against various brain injuries and antiapoptotic functions in several tissue types .

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.