Expression System: E. coli-derived, ensuring cost-effective production of non-glycosylated protein .
Formulation: Lyophilized powder in phosphate-buffered saline (PBS), requiring reconstitution in deionized water (0.5 mg/mL working concentration) .
Storage: Stable at -20°C in lyophilized form; reconstituted protein retains activity for limited periods at 4°C .
TSLP signals through a heterodimeric receptor complex composed of:
Receptor assembly follows a sequential mechanism: TSLP first binds TSLPR, enabling recruitment of IL-7Rα .
Induces CD11c+ dendritic cell maturation, upregulating costimulatory molecules (CD40, CD80) and enhancing T cell priming .
Stimulates monocytes to release T cell-attracting chemokines (e.g., TARC, MDC) .
Drives naive T cells toward a pro-allergic phenotype (IL-4, IL-5, IL-13, TNF-α) while suppressing IL-10 and IFN-γ .
Implicated in allergic inflammation (asthma, atopic dermatitis) and autoimmune disorders .
Drug Development: Structural insights into TSLP’s helical bundle and receptor interface inform antibody design for allergic diseases .
Disease Models: Used to study CRLF2-rearranged B-cell acute lymphoblastic leukemia (BCP-ALL) and cytokine-driven inflammation .
Comparative Studies: Species-specific activity (no cross-reactivity between human and mouse TSLP) necessitates humanized models for preclinical testing .
Human Thymic Stromal Lymphopoietin (TSLP) is a member of the IL-2 cytokine family and a distant paralog of IL-7, initially discovered in mice as a factor supporting B cell development. The human homolog shares only 43% amino acid identity with mouse TSLP, yet maintains significant functional homology . Human TSLP consists of a four-helix bundle structure containing six conserved cysteine residues and multiple potential sites for N-linked glycosylation . Despite these structural differences, both human and murine TSLP signal through similar receptor complexes and activate overlapping pathways, though with species-specific variations in cellular responses and signaling intensity.
The functional TSLP receptor consists of two subunits: the TSLP receptor (TSLPR) chain and the IL-7 receptor α (IL-7Rα) chain . This heterodimeric receptor is expressed on various hematopoietic cell populations including T cells, B cells, NKT cells, monocytes/macrophages, basophils, and dendritic cells, as well as some non-hematopoietic lineages such as epithelial cells . The TSLPR subunit is structurally unique among hematopoietin receptors, containing a modified WSXWS motif and lacking a Box2 motif that is typically involved in Janus kinase (JAK) binding . This unique structure contributes to TSLP's specific signaling properties.
TSLP binding to its receptor activates multiple signaling cascades. Recent studies have demonstrated robust and sustained activation of JAK-1 and JAK-2 following TSLP signaling in primary human dendritic cells and primary human and mouse CD4+ T cells . Unlike IL-7 signaling (which utilizes JAK-1 and JAK-3), TSLP signaling employs the TSLPR-associated JAK-2 in concert with IL-7Rα-associated JAK-1. In humans, TSLP stimulation activates STATs 1, 3, 4, 5, and 6, as well as JAKs 1 and 2 . This activation pattern differs somewhat from mouse models, possibly due to cell type-specific differences in signaling mechanisms.
When producing His-tagged human TSLP, researchers should consider the following expression system characteristics:
Expression System | Advantages | Limitations | Recommended for |
---|---|---|---|
Mammalian (HEK293, CHO) | Proper folding, glycosylation | Higher cost, lower yield | Functional studies, bioassays |
Insect cells | Post-translational modifications, higher yield | Medium cost | Structural studies, functional assays |
E. coli | Low cost, high yield | Limited folding, no glycosylation | Structural studies, antibody production |
Cell-free systems | Rapid production | Lower yield | Initial testing, pilot experiments |
For studies requiring fully functional human TSLP, mammalian expression systems are generally preferred as they ensure proper protein folding and post-translational modifications.
Bioactivity validation of His-tagged human TSLP should include multiple complementary approaches:
Dendritic cell activation assay: Measure upregulation of costimulatory molecules (CD80, CD86) and production of Th2-promoting chemokines after TSLP stimulation .
Phosphorylation analysis: Assess JAK/STAT pathway activation, particularly phosphorylation of STAT5, in TSLP-responsive cells using Western blot or flow cytometry .
T cell differentiation assays: Evaluate the ability of TSLP-treated dendritic cells to prime naïve CD4+ T cells toward a Th2 phenotype .
Dose-response experiments: Determine EC50 values across different cell types and compare with untagged TSLP to ensure the His-tag doesn't interfere with function.
Receptor binding studies: Confirm binding affinity to recombinant TSLPR/IL-7Rα complexes using surface plasmon resonance or similar techniques.
When working with His-tagged human TSLP, researchers should address several technical considerations:
Tag position effects: N-terminal versus C-terminal His-tags may differently affect TSLP bioactivity; comparative testing is recommended.
Storage stability: Optimize buffer conditions (pH, salt concentration, additives) to maintain activity during freeze-thaw cycles.
Endotoxin contamination: Ensure rigorous endotoxin removal, as contamination can confound immunological experiments.
Species specificity: Remember that human TSLP does not cross-react with mouse TSLPR due to only 35% homology between receptors .
Concentration standardization: Establish protein concentration using multiple methods (Bradford, BCA, absorbance) to ensure accurate dosing in experiments.
Dendritic cells (DCs) are a major TSLP-responsive cellular subset in both humans and mice . Experimental applications include:
DC maturation studies: TSLP treatment induces a specific DC phenotype characterized by upregulation of costimulatory molecules while producing limited amounts of proinflammatory cytokines .
T cell priming: TSLP-activated DCs promote naïve CD4+ T cells to produce IL-4, IL-5, IL-13, and TNF, but not IL-10 or IFN-γ, creating a unique inflammatory Th2 phenotype .
Memory T cell maintenance: TSLP-activated DCs support CRTH2+ Th2 effector memory cells, suggesting a role in chronic allergic inflammation .
IgA production: TSLP-conditioned DCs augment epithelial cell-mediated IgA2 class switching through APRIL induction, relevant for mucosal immunity .
Tolerogenic function: Some studies suggest TSLP may induce tolerogenic DCs that drive regulatory T cell differentiation, though contradictory findings exist .
Humanized mouse models for studying human TSLP include:
Stromal cell-based model: Human bone marrow stroma cell line (HS27) transduced to secrete human TSLP is injected intraperitoneally into NSG mice, producing serum TSLP levels comparable to human blood .
Transgenic approaches: Potential models include NSG mice with human BAC of TSLP or "knock-in" of human TSLP into the mouse TSLP locus .
Each model has advantages and limitations:
Model Type | Advantages | Limitations | Research Applications |
---|---|---|---|
Stromal cell-based | Modifiable TSLP levels, physiological concentrations | Laborious (weekly injections), non-physiological production site | Studying B-cell lymphopoiesis, leukemia biology |
Transgenic (proposed) | More physiological expression, normal regulation | More complex to develop, fixed expression pattern | Long-term studies, developmental effects |
The stromal cell-based model has demonstrated that human TSLP increases human B-cell lymphopoiesis in NSG mice transplanted with CD34+ cells from human cord blood .
TSLP plays multiple roles in allergic diseases:
Atopic dermatitis (AD): Elevated TSLP expression is found in lesional skin of AD patients . In mice, overexpression of TSLP in skin induces AD-like features including epidermal thickening, dermal inflammatory infiltrates, and Th2 responses .
Asthma: Increased TSLP expression is detected in lung epithelium and bronchoalveolar lavage from asthmatics . TSLP expression correlates directly with Th2 cytokine levels and inversely with lung function .
Genetic associations: Multiple SNPs at the TSLP genomic locus across various ethnic backgrounds are associated with asthma susceptibility or protection . A large meta-analysis of North American genome-wide association studies identified TSLP as one of five genes reaching statistical significance for asthma association .
Atopic march: TSLP may contribute to the progression from AD to asthma and allergic rhinitis . Models suggest that TSLP's primary role may be establishing allergen-specific Th2 responses during initial sensitization .
Important species-specific differences exist in TSLP biology:
Feature | Human TSLP | Mouse TSLP | Research Implications |
---|---|---|---|
Amino acid identity | - | 43% homology to human | Limited cross-reactivity |
Receptor homology | - | 35% homology to human | No cross-species binding |
STAT activation | STATs 1, 3, 4, 5, 6 | Initially thought more limited | Different downstream effects |
JAK utilization | JAK1, JAK2 | Similar to human | Conserved signaling mechanism |
Cell type responses | Broad effects | Similar but not identical | Cell-specific differences |
These differences necessitate caution when extrapolating findings from mouse models to human disease. The lack of cross-reactivity between species means that human TSLP must be expressed in mouse models to study its function in vivo .
When investigating TSLP's role in leukemias, particularly B-cell precursor acute lymphoblastic leukemias (BCP-ALLs) with abnormal TSLP receptor expression, researchers face several challenges:
Receptor heterogeneity: Up to two-thirds of BCP-ALL in children with Down syndrome and 5-10% of BCP-ALL in children and adults without Down syndrome have acquired genomic aberrations leading to increased CRLF2 (TSLPR) expression .
Mutation context: TSLPR overexpression is often accompanied by additional somatic activating mutations in the receptor or downstream JAK signaling molecules .
Disease heterogeneity: Limited studies (n=2 primografts) have shown variable effects of human TSLP on leukemic cell growth .
Gene expression changes: While TSLP may not affect leukemic cell growth in some models, it can alter gene expression patterns to more closely resemble original patient samples, including increased expression of mTOR signaling genes associated with chemoresistance .
Microenvironment interactions: Local TSLP production in the bone marrow may protect residual leukemic cells from chemotherapy, suggesting a need for models that recapitulate both systemic and local TSLP effects .
Researchers encountering contradictory data on TSLP function should consider:
Context-dependent effects: TSLP can promote either inflammatory or tolerogenic responses depending on:
Cellular context (cell types present)
Disease phase (sensitization vs. challenge)
Concentration and timing of exposure
Local vs. systemic effects
Cell type-specific targeting: During sensitization, TSLP acts primarily on dendritic cells to drive Th2 priming of CD4+ T cells, while during challenge phases, CD4+ T cells rather than DCs require TSLP responsiveness .
Species differences: Contradictions may arise from extrapolating between mouse and human systems without accounting for species-specific biology.
Technical considerations: Variations in recombinant protein quality, experimental conditions, and detection methods can contribute to divergent results.
Biological redundancy: In some contexts, TSLP effects may be compensated by other cytokines or signaling pathways.
TSLP-targeted therapeutic approaches include:
Monoclonal antibodies: Humanized anti-TSLP antibodies have completed Phase I clinical trials for asthma .
Receptor antagonists: Molecules designed to block TSLP-receptor interactions.
Pathway inhibitors: Compounds targeting downstream signaling components like JAK-STAT pathways.
The clinical development of anti-TSLP therapeutics represents the translation of basic research findings into potential treatments for allergic disorders .
Emerging approaches to study human TSLP include:
Improved humanized mouse models:
Local tissue microenvironment studies:
Single-cell approaches:
Characterization of heterogeneous responses to TSLP at single-cell resolution
Identification of novel TSLP-responsive cell populations
Systems biology integration:
Network analysis of TSLP interactions with other epithelial-derived cytokines
Computational modeling of context-dependent responses
Key questions for future research include:
Physiological role: What is the normal function of TSLP in human development and homeostasis beyond its pathological roles?
Genetic influences: How do genetic variants in TSLP and its receptor contribute to disease risk or protection across populations?
Signaling complexity: What determines whether TSLP promotes inflammatory or tolerogenic responses in different contexts?
Cancer biology: How does TSLP contribute to leukemia progression and chemoresistance, and could TSLP-targeted therapies benefit these patients?
Bone marrow niche: What is the role of local TSLP production in the bone marrow microenvironment for normal and malignant hematopoiesis?
Therapeutic optimization: What biomarkers could predict response to TSLP-targeted therapies, and how should treatment be timed relative to disease progression?
For optimal stability and activity of His-tagged human TSLP, researchers should consider:
Parameter | Recommendation | Rationale |
---|---|---|
Storage temperature | -80°C for long-term; -20°C for working aliquots | Minimize freeze-thaw cycles |
Buffer composition | PBS or HEPES with 0.1% BSA or HSA | Prevent protein adsorption |
pH range | 7.0-7.4 | Maintain physiological conditions |
Freeze-thaw cycles | Limit to 5 or fewer | Prevent denaturation |
Working concentration | Prepare fresh dilutions from stock | Ensure accurate dosing |
Carrier protein | Consider adding 0.1-0.5% BSA | Prevent binding to plastic |
Stability testing should be performed before beginning extensive experimental series to ensure consistent bioactivity throughout the study period.
Critical controls for TSLP experiments include:
Tag-only control: Purified His-tag peptide to control for tag-specific effects.
Heat-inactivated TSLP: Heat-denatured protein to demonstrate specificity of biological effects.
Receptor blocking: Anti-TSLPR antibodies to confirm receptor dependence of observed effects.
Species controls: When using humanized models, include human and mouse TSLP comparisons to demonstrate species specificity .
Signaling pathway inhibitors: JAK/STAT inhibitors to confirm mechanism of action.
Positive controls: Well-characterized TSLP-responsive cell lines with established readouts.
Concentration range: Full dose-response curves to identify optimal working concentrations.
His-tagged human TSLP can be employed in various in vitro models of allergic inflammation:
Dendritic cell-T cell co-culture systems:
Culture human monocyte-derived DCs with His-tagged TSLP (5-10 ng/ml) for 24 hours
Co-culture with naïve CD4+ T cells to assess Th2 polarization
Measure cytokine production (IL-4, IL-5, IL-13) and surface marker expression
Reconstituted human epithelial models:
Apply environmental allergens to airway or skin epithelial cultures
Measure TSLP production and compare to exogenous His-tagged TSLP effects
Assess epithelial barrier function and inflammatory mediator production
Mast cell and basophil activation:
Prime cells with IgE, then stimulate with His-tagged TSLP alone or in combination with allergens
Measure degranulation and cytokine production
Compare responses between healthy donors and allergic patients
TSLP plays complex roles in B-cell biology and leukemia:
His-tagged human TSLP enables precise study of these effects through:
Dose-response experiments to characterize hypersensitivity
Pull-down assays to identify binding partners
Proximity labeling to map signaling complexes
Competition assays with untagged TSLP or receptor antagonists
When investigating TSLP's contribution to the atopic march (progression from atopic dermatitis to asthma and allergic rhinitis), researchers should consider:
Sequential exposure protocols:
Initial skin sensitization with allergen and TSLP
Subsequent airway challenge with allergen alone
Assessment of airway inflammation and hyperresponsiveness
Tissue-specific TSLP manipulation:
Models with inducible TSLP expression in specific tissues
Comparison of local vs. systemic TSLP effects
Timing considerations:
Different time intervals between sensitization and challenge
Age-dependent effects relevant to pediatric progression
Cell tracking approaches:
Fate mapping of allergen-specific T cells from skin to airways
Identification of migratory dendritic cell populations
Role of TSLP in different phases:
Current humanized mouse models for studying human TSLP have limitations, including laborious maintenance requirements and failure to recapitulate normal TSLP production sites and regulation . Future advances could include:
Transgenic approaches:
Inducible systems:
Tissue-specific and temporally controlled TSLP expression
Models that can switch between human and mouse TSLP expression
Multi-cytokine humanization:
Combined expression of human TSLP with other human cytokines
More complete recapitulation of human immune microenvironments
Patient-derived xenograft models:
These advances would help resolve fundamental questions about TSLP biology and accelerate translation of findings to clinical applications.
Future development of TSLP-targeted therapeutics may focus on:
Selective targeting:
Tissue-specific delivery of TSLP inhibitors
Targeting specific downstream signaling pathways
Combination approaches:
TSLP blockade combined with inhibition of other allergic mediators
Sequential or alternating therapeutic regimens
Personalized medicine:
Biomarker-guided patient selection for TSLP-targeted therapies
Genetic profiling to identify optimal responders
Novel indications:
Timing optimization:
Intervention at specific disease stages for maximum efficacy
Prevention strategies for high-risk individuals
Human TSLP research represents a rapidly evolving field with significant implications for understanding and treating allergic diseases, certain leukemias, and potentially other inflammatory conditions.
Thymic Stromal Lymphopoietin (TSLP) is a hemopoietic cytokine that plays a crucial role in the immune system. It signals through a heterodimeric receptor complex composed of the thymic stromal lymphopoietin receptor (TSLPR) and the Interleukin-7 receptor alpha chain (IL-7Rα). TSLP is primarily expressed in the heart, liver, and prostate, and it is involved in the regulation of immune responses, particularly in allergic inflammation .
The human recombinant TSLP with a His tag is typically produced in Escherichia coli (E. coli). The recombinant protein is a single, non-glycosylated polypeptide chain containing 141 amino acids, including a 10 amino acid His tag at the N-terminus. The total calculated molecular mass is approximately 16.2 kDa . The protein is filtered and lyophilized in phosphate-buffered saline for storage and use .
TSLP impacts myeloid cells and induces the release of T cell-attracting chemokines from monocytes. It also promotes the growth of CD11c(+) dendritic cells. TSLP-activated dendritic cells prime naive T cells to produce pro-allergic cytokines such as Interleukin-4 (IL-4), Interleukin-5 (IL-5), Interleukin-13 (IL-13), and TNF-alpha, while down-regulating Interleukin-10 (IL-10) and IFN-gamma .
TSLP functions as an alarmin and growth factor in the immune system. It is involved in type 2 immune responses (TH2 responses) and the maturation and recruitment of dendritic cells, T cells, B cells, neutrophils, mast cells, and other lymphoid cells. TSLP can be produced by epithelial and stromal cells in the lung, skin, and gastric system, as well as by dendritic cells, basophils, and mast cells . Its expression can be induced by infections, pro-inflammatory cytokines, proteases, and mechanical injury .
TSLP is linked to allergic reactions such as asthma, atopic dermatitis, and food allergies. It induces the expression of OX40L, CD80, and CD86, and stimulates CD4+ T cells . In 2021, the TSLP-neutralizing antibody tezepelumab was approved for the treatment of severe asthma . Targeting TSLP is an active area of investigation, with ongoing clinical trials for the treatment of autoimmune disorders .