Engineered from HEK293 cells, these reporter lines enable detection of bioactive TPO via STAT5-dependent SEAP (secreted embryonic alkaline phosphatase) expression :
Cell Proliferation: Recombinant murine TPO stimulates MO7e megakaryocytic cells with an ED<sub>50</sub> of ≤4–5 ng/ml .
Platelet Regulation: Enhances megakaryocyte differentiation, platelet aggregation, and adhesion .
Drug Screening: HEK-Blue™ TPO cells identify anti-TPO antibodies or inhibitors targeting the JAK2/STAT5 pathway .
Disease Models: JAK2 V617F mutations in myeloproliferative neoplasms downregulate TPO-R levels, reversible via JAK2/proteasome inhibitors .
Recombinant murine TPO produced in HEK293 cells undergoes rigorous validation:
Thrombopoietin (TPO), also known as MGDF, is a glycoprotein hormone primarily produced in the liver and kidneys. It plays a crucial role in platelet production by stimulating the production and differentiation of megakaryocytes, specialized bone marrow cells responsible for platelet formation. TPO is essential for megakaryocytopoiesis, the process of megakaryocyte proliferation and maturation, and overall platelet production (thrombopoiesis).
Recombinant Mouse TPO, expressed in HEK293 cells, is a non-glycosylated polypeptide chain consisting of 341 amino acids (22-356 a.a). With a molecular weight of 36.4 kDa, this TPO variant has a 6 amino acid His-tag fused at its C-terminus. Purification is achieved through proprietary chromatographic techniques.
The TPO protein solution has a concentration of 0.5mg/ml and is prepared in Phosphate-Buffered Saline (pH 7.4) with 10% glycerol.
For short-term storage (2-4 weeks), keep refrigerated at 4°C. For extended storage, freeze at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Repeated freezing and thawing should be avoided.
Purity is determined using a cell proliferation assay with MO7e human megakaryocytic leukemic cells. The ED50 is approximately 4 ng/ml.
Biological activity is assessed by the ability to agglutinate human red blood cells, with an ED50 greater than or equal to 25 µg/ml.
Thpo, C-mpl ligand,ML, Megakaryocyte colony-stimulating factor, Megakaryocyte growth and development factor, MGDF, Myeloproliferative leukemia virus oncogene ligand, Mgdf, Ml, Mpllg, Tpo, thrombopoietin isoform 1.
HEK293.
SPVAPACDPR LLNKLLRDSH LLHSRLSQCP DVDPLSIPVL LPAVDFSLGE WKTQTEQSKA QDILGAVSLL LEGVMAARGQ LEPSCLSSLL GQLSGQVRLL LGALQGLLGT QLPLQGRTTA HKDPNALFLS LQQLLRGKVR FLLLVEGPTL CVRRTLPTTA VPSSTSQLLT LNKFPNRTSG LLETNFSVTA RTAGPGLLSR LQGFRVKITP GQLNQTSRSP VQISGYLNRT HGPVNGTHGL FAGTSLQTLE ASDISPGAFN KGSLAFNLQG GLPPSPSLAP DGHTPFPPSP ALPTTHGSPP QLHPLFPDPS TTMPNSTAPH PVTMYPHPRN LSQETHHHHH H.
TPO (thrombopoietin) is a key hematopoietic growth factor that plays a vital role in the regulation of megakaryocytopoiesis and the maintenance of hematopoietic stem cells (HSCs) . For research purposes, specialized reporter systems such as HEK-Blue™ TPO cells have been engineered from human embryonic kidney 293 cell lines to detect bioactive TPO by monitoring the activation of the JAK2/STAT5 pathway . These cells express the human TPO receptor (TPO-R, aka CD110), human STAT5b, and a STAT5-inducible secreted embryonic alkaline phosphatase (SEAP) reporter . When TPO binds to its receptor, it triggers signaling that leads to STAT5 activation and subsequent SEAP production, which can be readily assessed using QUANTI-Blue™ Solution .
HEK-Blue TPO cells are capable of detecting both human and murine TPO, although with different sensitivity ranges :
Parameter | Specification |
---|---|
Detection range for human TPO | 3 - 100 ng/ml |
Detection range for murine TPO | 0.1 - 10 ng/ml |
Antibiotic resistance | Blasticidin, Hygromycin B, Zeocin® |
Cross-reactivity | Responds to human and murine TPO |
Additional reactivity | Responds to IFN-α, IFN-β, and IFN-γ |
Non-responsive to | Human IL-2 and human IL-15 |
This dual-species responsiveness makes these cells versatile tools for comparative studies between human and mouse TPO biology . The higher sensitivity to murine TPO (detectable from 100 pg/ml) compared to human TPO (detectable from 3 ng/ml) should be considered when designing cross-species experiments .
While TPO research focuses on hematopoiesis, Tryptophan hydroxylase type 2 (Tph2) represents a separate but equally important research area focused on serotonin (5-HT) biosynthesis in the brain . Tph2 is the rate-limiting enzyme for serotonin production in the central nervous system, and its dysfunction can lead to 5-HT deficiency with potential behavioral implications . Studies have demonstrated that the construct pIRES-hrGFP-1a-Tph2-FLAG can enhance 5-HT production when transfected into HEK-293 cells (observed 48 hours post-transfection) and when administered to mice (observed three days after ocular administration in the hypothalamus and amygdala, but not in the brainstem) . This research path connects TPO and HEK studies to broader neuropsychiatric research, as 5-HT deficiency has been implicated in conditions including alcoholism, impulsive behavior, anxiety, and depression .
The following protocol details the methodological approach for detecting TPO activity with HEK-Blue TPO cells :
Day 1:
Cell preparation: Gently rinse cells twice with pre-warmed PBS; detach cells in PBS for 2-3 min at 37°C (avoid trypsin as it may alter receptor expression); prepare cell suspension at ~280,000 cells/ml
Sample loading: Add 20 μl of sample per well in a flat-bottom 96-well plate
Controls: Add 20 μl of positive control (recombinant human TPO at 10 ng/ml) and negative control (recombinant human IL-2 at 10 ng/ml) in separate wells
Cell addition: Add 180 μl of HEK-Blue TPO cell suspension (~50,000 cells) per well
Incubation: Incubate overnight at 37°C in 5% CO2
Day 2:
6. QUANTI-Blue preparation: Prepare QUANTI-Blue Solution according to manufacturer's instructions
7. Detection setup: Add 20 μl of induced HEK-Blue TPO cells supernatant to 180 μl of QUANTI-Blue Solution
8. Incubation: Incubate at 37°C for 30 min to 3 hours
9. Measurement: Determine SEAP levels using a spectrophotometer at 620-655 nm
Critical considerations include avoiding trypsin for cell detachment as prolonged trypsin action may alter cell surface receptor expression, maintaining cells at 70-80% confluency, and not allowing growth to 100% confluency .
Distinguishing between TPO-specific signaling and non-specific activation requires careful experimental controls. HEK293 cells endogenously express receptors for type I and type III interferons (IFNs), making them responsive to IFN-α, IFN-β, and IFN-γ . The response profile to various cytokines has been characterized:
Cytokine | Typical Concentration | Response in HEK-Blue TPO cells |
---|---|---|
Human TPO | 10 ng/ml | Strong positive |
Murine TPO | 10 ng/ml | Strong positive |
Human IFN-α | 30 U/ml | Positive |
Human IFN-β | 30 U/ml | Positive |
Human IFN-γ | 10 ng/ml | Positive |
Human IL-2 | 10 ng/ml | Negative |
Human IL-15 | 10 ng/ml | Negative |
To ensure signaling specificity, researchers should include appropriate negative controls (IL-2, IL-15) and be aware of potential activation through endogenous IFN receptors . Additional validation can come from dose-response experiments showing characteristic EC50 values within the expected ranges for human and murine TPO .
Several factors can influence TPO-R expression on the cell surface in experimental settings, which directly impacts assay sensitivity and reproducibility:
Cell passage number: HEK-Blue TPO cells are guaranteed stable for 20 passages following thawing
Confluency: Cells should be maintained at 70-80% confluency and not allowed to reach 100% confluency
Cell detachment methods: Prolonged trypsin exposure may alter receptor expression; gentle physical detachment in PBS is recommended
Growth conditions: Cells require specific growth medium with selection antibiotics (blasticidin, hygromycin B, Zeocin®)
Cell surface verification: Human TPO-R surface expression should be periodically verified by flow cytometry to ensure consistent receptor levels
The validation data for HEK-Blue TPO cells confirms human TPO-R surface expression through flow cytometry and STAT5 expression through RT-qPCR , providing important quality control benchmarks for researchers.
When studying TPO signaling pathways, a comprehensive control strategy should include:
Positive controls: Recombinant human TPO (10 ng/ml) or murine TPO within their respective detection ranges (3-100 ng/ml for human TPO; 0.1-10 ng/ml for murine TPO)
Negative controls: Human IL-2 and IL-15 (10 ng/ml each), which do not activate STAT5 through TPO-R
Cross-reactivity controls: IFN-α (30 U/ml), IFN-β (30 U/ml), and IFN-γ (10 ng/ml) to account for endogenous receptor activation
Antibody controls: Anti-TPO or anti-TPO-R antibodies can be screened to confirm specificity of the signaling pathway
Dose-response curves: Full concentration ranges should be tested to establish proper EC50 values for comparison with published standards
This control framework enables researchers to distinguish specific TPO signaling from background or non-specific activation, ensuring experimental rigor.
When faced with inconsistencies between HEK cell assays and mouse models, researchers should consider several methodological approaches:
Physiological context assessment: HEK-Blue TPO cells provide a controlled environment focused solely on receptor-ligand interactions and immediate signaling, while in vivo models incorporate complex physiological contexts including pharmacokinetics, tissue distribution, and cross-talking signaling pathways
Parameter alignment: Ensure that comparable parameters are being measured across systems; SEAP reporter activity in vitro may not directly correlate with physiological endpoints in vivo
Bridging experiments: Use primary cells derived from mice in parallel with HEK cell assays under identical conditions to identify system-specific differences
Time-course considerations: In vitro responses (typically measured after overnight incubation) may have different kinetics compared to in vivo responses (which may take days to develop fully)
Genetic validation: For Tph2 studies, implementing the same genetic construct (e.g., pIRES-hrGFP-1a-Tph2-FLAG) in both HEK-293 cells and mouse models provides a more direct comparison
Translating TPO concentrations between in vitro HEK cell systems and in vivo mouse models requires consideration of several factors:
Detection range differences: HEK-Blue TPO cells have defined detection ranges (3-100 ng/ml for human TPO; 0.1-10 ng/ml for murine TPO) , while physiological TPO levels in mice may differ
Bioavailability factors: In vivo administration must account for absorption, distribution, metabolism, and excretion, which do not affect in vitro systems
Receptor density variations: TPO-R expression levels may differ between engineered HEK cells and native target cells in mice
Biological amplification: In vivo systems may demonstrate signal amplification through downstream pathways not present in the HEK reporter system
Experimental validation: Pilot dose-finding studies in mice should establish dose-response relationships that can then be correlated with in vitro EC50 values
These considerations help researchers develop more physiologically relevant experimental designs that bridge the gap between cell culture and animal models.
For rigorous analysis of TPO dose-response data, researchers should employ these statistical approaches:
Nonlinear regression: Four-parameter logistic regression is ideal for TPO dose-response curves, generating EC50 values, Hill slopes, and maximum response parameters
Replicate design: Technical replicates (minimum triplicates) and biological replicates across independent experiments provide robust statistical power
Normalization strategies: Raw OD values at 630 nm can be normalized to percent maximum response or fold-change over baseline to facilitate comparison between experiments
Statistical comparison methods: ANOVA with appropriate post-hoc tests for comparing multiple concentrations or conditions; t-tests for comparing specific EC50 values between experimental groups
Graphical representation: Log-scale for TPO concentration with error bars representing standard error of mean (SEM) provides clear visualization of dose-response relationships
Proper statistical analysis ensures that subtle differences in TPO activity or receptor function can be reliably detected and interpreted.
STAT5 activation patterns in TPO signaling provide important molecular insights that should be interpreted in context:
Pathway specificity: STAT5 activation through TPO-R indicates specific engagement of the JAK2/STAT5 pathway, a key mediator of TPO's biological effects
Activation kinetics: The time-course of STAT5 activation (measured via SEAP production) reflects receptor-proximal signaling events that precede biological responses
Dose-dependency interpretation: EC50 values for STAT5 activation correlate with receptor binding affinity and signaling efficiency, allowing comparison between different TPO variants or mimetics
Cross-talk assessment: Activation by both TPO and interferons indicates potential pathway integration or compensatory mechanisms that may be physiologically relevant
Biological correlation: STAT5 activation in vitro should be correlated with known downstream biological effects such as cell proliferation, differentiation, or survival in appropriate cellular contexts
Understanding these molecular signaling patterns helps researchers connect biochemical observations to biological significance in both research and therapeutic applications.
To differentiate 5-HT increases resulting specifically from Tph2 overexpression versus other mechanisms, researchers should implement these methodological controls:
Vector controls: Compare pIRES-hrGFP-1a-Tph2-FLAG with empty vector controls to isolate effects specific to Tph2 overexpression
Enzymatic inhibition: Apply Tph2-specific inhibitors to confirm that observed 5-HT increases depend on Tph2 enzymatic activity
Temporal correlation: Establish the time-course relationship between Tph2 expression (measured by protein levels) and subsequent 5-HT increases
Regional specificity: Compare 5-HT changes across brain regions with different levels of transgene expression (e.g., the study showed increases in hypothalamus and amygdala but not brainstem)
Substrate availability assessment: Measure tryptophan levels to ensure that 5-HT increases are not limited by substrate availability rather than enzyme activity
Biochemical validation: Use LC-MS/MS or other sensitive analytical techniques to directly measure 5-HT and its metabolites, distinguishing new synthesis from altered metabolism or reuptake
These approaches ensure that observed phenotypes can be confidently attributed to the specific molecular mechanism under investigation.
HEK-Blue TPO cells provide a powerful platform for screening TPO mimetics and receptor agonists through several methodological approaches:
High-throughput screening: The colorimetric SEAP reporter system allows rapid evaluation of large compound libraries in 96-well or 384-well formats
Comparative potency assessment: Side-by-side comparison with recombinant TPO establishes relative potency (EC50) and efficacy (maximum response) of candidate molecules
Structure-activity relationship studies: Systematic modification of lead compounds can be rapidly evaluated to optimize binding and signaling properties
Antibody screening: Both anti-TPO and anti-TPO-R antibodies can be assessed for agonist or antagonist activity
Pathway selectivity profiling: Comparison of STAT5 activation with activation of other pathways can identify biased agonists with potential therapeutic advantages
The system's ability to detect both human and murine TPO makes it particularly valuable for developing therapeutics with cross-species activity, facilitating translation from preclinical to clinical studies .
Emerging research directions are exploring connections between TPO signaling and Tph2 function, opening new avenues for integrated hematopoietic-neurological studies:
Shared signaling pathways: Both TPO (via JAK2/STAT5) and serotonin receptors activate overlapping intracellular pathways, suggesting potential cross-talk mechanisms
Platelet-serotonin interactions: Platelets (regulated by TPO) are major carriers of peripheral serotonin (produced via Tph1, related to Tph2), creating a functional link between these systems
Stem cell regulation: Both TPO and serotonin influence stem cell niches, with potential cooperative effects on hematopoietic stem cell maintenance and differentiation
Genetic engineering approaches: Methods used to express Tph2 in HEK cells and mouse models can be adapted to study TPO pathway components in neurological contexts
Therapeutic convergence: Understanding how these systems interact could lead to novel therapeutic approaches for conditions affecting both hematopoietic and neurological functions
This integrative research direction acknowledges the complex interplay between hematopoietic factors and neurotransmitter systems in health and disease.
Several technological innovations are advancing TPO research across both in vitro and in vivo systems:
Enhanced reporter systems: Next-generation reporters beyond SEAP, including luminescent and fluorescent options, enable real-time monitoring of TPO signaling kinetics with higher sensitivity
CRISPR/Cas9 genome editing: Precise modification of TPO-R, JAK2, STAT5, or downstream components in both HEK cells and mouse models facilitates mechanistic studies of specific signaling nodes
Single-cell analysis: Application of single-cell technologies to TPO-responsive populations reveals heterogeneity in signaling responses not captured by bulk assays
Organoid systems: Development of megakaryocyte organoids provides a bridge between 2D cell culture and in vivo models, incorporating tissue architecture and cell-cell interactions
Non-viral gene transfer: Methods demonstrated for Tph2 expression (using pIRES-hrGFP-1a-Tph2-FLAG) can be adapted for modulating TPO pathway components in vivo without viral vectors
Computational modeling: Integration of in vitro signaling data with in vivo responses enables predictive modeling of TPO effects across different physiological contexts
These technological advances are accelerating both basic research on TPO biology and translational efforts to develop TPO-targeted therapeutics for hematological disorders.
Thrombopoietin (TPO) is a crucial cytokine involved in the regulation of platelet production. It primarily influences the proliferation and maturation of megakaryocytes, the bone marrow cells responsible for producing platelets. Recombinant forms of TPO, such as Mouse Recombinant Thrombopoietin expressed in Human Embryonic Kidney (HEK) cells, are widely used in research and therapeutic applications.
Recombinant Mouse Thrombopoietin (HEK) is typically produced in HEK293 cells, a human cell line derived from embryonic kidney cells. This recombinant protein is a single, non-glycosylated polypeptide chain containing 341 amino acids, with a molecular mass of approximately 36.4 kDa . The protein is often purified using proprietary chromatographic techniques to ensure high purity and biological activity.
Thrombopoietin plays a vital role in hematopoiesis, particularly in the production of platelets. It binds to its receptor, c-Mpl, on the surface of megakaryocytes and their progenitors, promoting their proliferation and differentiation . This cytokine is also essential for the maintenance of hematopoietic stem cells (HSCs), ensuring a steady supply of blood cells throughout an organism’s life.
Recombinant Mouse Thrombopoietin (HEK) is used in various research applications, including:
Recombinant Mouse Thrombopoietin (HEK) is usually supplied in a lyophilized form to ensure stability during storage and transport. It can be reconstituted in phosphate-buffered saline (PBS) and stored at -80°C for long-term use . Proper storage and handling are essential to maintain the protein’s biological activity and prevent degradation.