IL 7 Rat

Interleukin-7 Rat Recombinant
Shipped with Ice Packs
In Stock

Description

Biological Functions in Rat Immune Systems

IL-7 is indispensable for:

  • Lymphopoiesis: Driving hematopoietic stem cell differentiation into lymphoid progenitors (T, B, and NK cells) .

  • T Cell Homeostasis: Enhancing survival and proliferation of naïve and memory T cells via Bcl-2 upregulation and PUMA suppression .

  • Immune Reconstitution: Accelerating T cell recovery post-chemotherapy or bone marrow transplantation .

Key Mechanism: IL-7 activates JAK/STAT5 and PI3K/AKT pathways, promoting cell-cycle progression (via cyclin D1) and inhibiting apoptosis (via Bcl-2) .

Table 1: Experimental Applications of Rat IL-7

Study FocusModelKey OutcomeReference
Diabetic Wound HealingStreptozotocin-induced diabetic ratsIL-7-transfected MSCs enhanced angiogenesis, wound closure, and VEGF/HGF expression .
Myocardial I/R InjuryRat cardiac I/R modelAnti-IL-7 antibody reduced infarct size, troponin T levels, and macrophage-driven apoptosis .
Glioma TherapyRat glioma xenograftsIL-7 synergized with IFN-γ to boost antitumor CD8+ T cell activity .

Notable Results:

  • Wound Healing: IL-7 overexpression in mesenchymal stem cells (MSCs) increased vascular endothelial growth factor (VEGF) and hepatocyte growth factor (HGF), accelerating diabetic wound closure by 15 days .

  • Cardiac Injury: IL-7 knockout or anti-IL-7 antibody treatment reduced myocardial infarction area by 40% and improved cardiac function by modulating macrophage polarization .

  • Cancer Immunotherapy: IL-7 enhanced IFN-γ-mediated tumor regression in glioma models, correlating with elevated cytotoxic T cell activity .

Therapeutic and Biotechnological Applications

  • Recombinant IL-7: Used to expand pre-B cells and restore thymopoiesis in IL-7-deficient mice .

  • IL-7/Antibody Complexes: Demonstrated 50–100x higher bioactivity than free IL-7, driving T cell proliferation even in lymphoreplete conditions .

  • Gene Therapy: IL-7-transfected MSCs improved tissue repair in chronic wounds, highlighting clinical potential for regenerative medicine .

Mechanistic Insights and Pathological Roles

  • Autoimmunity: IL-7 promotes IFN-γ-producing Th1 and Tc1 cells, exacerbating type 1 diabetes in NOD mice. Anti-IL-7Rα antibodies reversed diabetes by suppressing pathogenic T cells .

  • Inflammation: IL-7 amplifies IL-17-producing γδ T cells, contributing to tissue damage in ischemia-reperfusion injury .

Comparative Analysis: Rat vs. Murine IL-7

While IL-7’s core functions are conserved across species, rat-specific findings include:

  • Unique angiogenic responses in diabetic wound models .

  • Differential macrophage polarization in cardiac injury compared to murine systems .

Challenges and Future Directions

  • Dosing Limitations: High IL-7 doses are required for therapeutic effects due to rapid clearance .

  • Dual Roles in Cancer: Pro-tumorigenic effects via lymphocyte survival vs. antitumor activity through CD8+ T cell activation .

Product Specs

Introduction
Interleukin-7 (IL-7) is a cytokine crucial for the development of B and T cells. In conjunction with hepatocyte growth factor (HGF), it forms a heterodimer that stimulates the growth of pre-pro-B cells. IL-7 is known to be a crucial cofactor for the V(D)J rearrangement of the T cell receptor beta (TCRB) during the early stages of T cell development. Its production locally by intestinal epithelial and epithelial goblet cells suggests a role in regulating intestinal mucosal lymphocytes. Studies with knockout mice models have indicated that IL-7 is essential for lymphoid cell survival.
Description
Recombinant Rat IL-7, produced in E. coli, is a single, non-glycosylated polypeptide chain comprising 129 amino acids. With a molecular weight of 15.0 kDa, this IL-7 undergoes purification using proprietary chromatographic techniques.
Physical Appearance
White, sterile-filtered, lyophilized (freeze-dried) powder.
Formulation
Lyophilized from a 0.2µm filtered solution concentrated in phosphate-buffered saline (PBS) at pH 7.4.
Solubility
For reconstitution, it is recommended to dissolve the lyophilized IL-7 in sterile 18 MΩ-cm H₂O to a concentration of at least 100µg/ml. This solution can be further diluted in other aqueous solutions.
Stability
Lyophilized IL-7, while stable at room temperature for 3 weeks, is best stored desiccated below -18°C. Upon reconstitution, IL-7 should be stored at 4°C for a period of 2-7 days. For long-term storage, it should be kept at -18°C. Avoid repeated freeze-thaw cycles.
Purity
Determined by SDS-PAGE analysis to be greater than 98.0%.
Biological Activity
The ED50, determined by the dose-dependent stimulation of murine 2E8 cell proliferation, is less than 0.2 ng/ml, which corresponds to a specific activity exceeding 5,000,000 IU/mg.
Synonyms
Lymphopoietin 1 (LP-1), pre-B cell factor, IL-7.
Source
Escherichia Coli.
Amino Acid Sequence
DCHIKDKDGK AFGSVLMISI NQLDKMTGTD SDCPNNEPNF FKKHLCDDTK EAAFLNRAAR KLRQFLKMNI SEEFNDHLLR VSDGTQTLVN CTSKEEKTIK EQKKNDPCFL KRLLREIKTC WNKILKGSI

Q&A

What are the fundamental roles of IL-7 in rat T cell development and homeostasis?

IL-7 serves as a critical cytokine that regulates multiple aspects of T cell biology in rats and other mammalian models. Unlike many cytokines, IL-7 is not produced by T cells themselves but primarily by non-lymphoid lineage stromal cells and epithelial cells with limited numbers. This creates a dependency where T cells rely on extrinsic IL-7, with the amount available in vivo acting as a major factor in maximizing and maintaining T cell populations in peripheral tissues .

The primary functions of IL-7 in rat T cell biology include:

  • Providing essential metabolic cues for T cell survival

  • Promoting the survival of both naïve and memory T cell populations

  • Maintaining functional fitness of T cells through various signaling pathways

  • Regulating T cell numbers through availability-based mechanisms

When designing experiments to study IL-7 function in rats, researchers should consider:

  • The source and glycosylation status of recombinant IL-7 used in experiments

  • Appropriate dosing based on the specific rat strain and experimental context

  • Complementary analysis of both survival and proliferation parameters

  • Assessment of downstream signaling molecules, particularly STAT5 phosphorylation and Bcl-2 expression

How does IL-7 receptor expression vary across different rat T cell subpopulations?

IL-7 receptor expression shows significant variation across T cell subsets in rats, with important functional implications. The IL-7 receptor consists of the IL-7Rα chain (CD127) paired with the common gamma chain (γc). Expression levels of IL-7Rα differ dramatically between:

  • Recent thymic emigrants (RTEs): Express lower levels of surface IL-7Rα

  • Mature naïve T cells: Express higher levels of IL-7Rα

  • Memory T cells: The CD44highIL-7Rαhigh population represents true memory cells

  • Effector T cells: Often characterized as CD44highIL-7Rαlow cells

Interestingly, despite lower IL-7Rα expression, RTEs demonstrate more efficient IL-7 signaling than mature naïve T cells, showing increased STAT5 phosphorylation in response to IL-7. This apparent paradox is resolved by understanding that RTEs interpret IL-7 signals differently, preferentially inducing anti-apoptotic proteins like Bcl-2 .

For accurate assessment of IL-7 receptor expression in rat models, researchers should:

  • Use multiparameter flow cytometry with validated antibodies for rat IL-7Rα

  • Include markers to precisely identify T cell developmental stages

  • Consider functional readouts alongside receptor expression analysis

  • Account for receptor downregulation after IL-7 exposure when designing experiments

How do signaling pathways differ between recent thymic emigrants and mature naïve T cells in response to IL-7?

Recent research has revealed striking differences in how recent thymic emigrants (RTEs) and mature naïve T cells respond to IL-7 stimulation. These differences represent a fundamental biological adaptation that influences T cell homeostasis:

ParameterRecent Thymic Emigrants (RTEs)Mature Naïve T Cells
IL-7Rα expressionLowerHigher
STAT5 phosphorylationMore efficientLess efficient
Bcl-2 inductionStrongerWeaker
Proliferative responseReducedEnhanced
Primary outcomeSurvivalProliferation

RTEs preferentially interpret IL-7 signals as survival cues through robust upregulation of the anti-apoptotic protein Bcl-2, which paradoxically inhibits proliferation. In contrast, mature naïve T cells show less Bcl-2 induction but greater proliferative responses to IL-7 .

This dichotomy has been demonstrated through adoptive transfer experiments in lymphopenic hosts, where:

  • Naïve T cells proliferated more efficiently than RTEs

  • CD8+ RTEs underwent cell proliferation at a slower rate than naïve T cells

  • Both populations showed minimal proliferation in IL-7-deficient hosts, confirming IL-7 dependency

  • These differences persisted even with increased IL-7 availability

These findings suggest that IL-7 responsiveness in RTEs is evolutionarily designed to maximize survival at the expense of reduced proliferation, consistent with RTEs serving as a T cell subpopulation rich in diversity but not in frequency .

What molecular mechanisms explain the differential effects of IL-7 on T cell survival versus proliferation?

The dual role of IL-7 in promoting both survival and proliferation is governed by distinct molecular mechanisms that can be differentially regulated in various T cell subsets:

Survival Mechanisms:

  • Upregulation of anti-apoptotic Bcl-2 family proteins, particularly Bcl-2 itself

  • Maintenance of mitochondrial integrity

  • Prevention of the activation of pro-apoptotic molecules

  • Metabolic support through glucose uptake and utilization

Proliferation Mechanisms:

  • Activation of cell cycle regulators

  • Modulation of cyclin-dependent kinases

  • Regulation of cell cycle inhibitory molecules

  • Cross-talk with TCR signaling components

The balance between these pathways appears to be developmentally regulated, with RTEs showing preferential activation of survival pathways through sustained Bcl-2 upregulation. This mechanistic distinction is critical for understanding how IL-7 therapy might differentially affect T cell populations in various disease contexts .

Experimentally, researchers can distinguish between these effects by:

  • Assessing Bcl-2 expression (primarily associated with survival)

  • Measuring proliferation markers like Ki-67 or BrdU incorporation

  • Analyzing cell cycle progression through flow cytometry

  • Comparing responses in different T cell developmental stages

What are optimal protocols for studying IL-7 effects in rat lymphopenia models?

Lymphopenia models provide a powerful system for studying IL-7 biology, as reduced T cell numbers increase per-cell IL-7 availability. For rat studies, researchers should consider the following methodological approaches:

Experimental Design Elements:

  • Model selection:

    • Surgical thymectomy followed by irradiation or chemotherapy

    • Genetic models (e.g., Rag-deficient rats if available)

    • Antibody-mediated depletion of T cells

  • Cell transfer experiments:

    • Purification of donor T cell populations using negative selection to avoid activation

    • Labeling cells with proliferation dyes (CFSE, CellTrace) to track divisions

    • Adopting transfer numbers that avoid competition for IL-7 (typically 1-5×10^6 cells)

    • Including both RTEs and mature naïve T cells for comparative analysis

  • Analysis timepoints:

    • Early (3-5 days): Primarily survival effects

    • Intermediate (7-14 days): Initial proliferation waves

    • Late (21+ days): Establishment of steady-state in reconstituted animals

  • IL-7 manipulation approaches:

    • Recombinant IL-7 administration (typically 50 μg/kg, s.c., weekly)

    • IL-7/anti-IL-7 complexes for extended half-life

    • Anti-IL-7Rα blocking antibodies

    • Genetic approaches to modulate IL-7 signaling components

From previous studies, we know that naïve CD8+ T cells show greater IL-7-dependent lymphopenia-induced proliferation than CD8+ RTEs, despite the latter showing more efficient IL-7 signaling. This observation has been confirmed through experiments in IL-7-deficient lymphopenic hosts, where both populations showed minimal proliferation .

How should researchers optimize IL-7 dosing and administration protocols in rat disease models?

Optimization of IL-7 dosing is critical for achieving desired immunological effects while minimizing potential side effects:

Dosing Considerations:

  • Based on primate studies, a starting dose of 50 μg/kg administered subcutaneously on a weekly schedule has shown efficacy in SIV models

  • Dose-response studies should be conducted to establish the minimum effective dose for each application

  • Duration of 7 weeks treatment has been effective in acute SIV infection models

  • Higher doses may be required for therapeutic versus prophylactic applications

Administration Routes:

  • Subcutaneous injection is most common and provides sustained release

  • Intravenous administration may be appropriate for acute interventions

  • Local delivery might be considered for tissue-specific applications

  • Novel delivery systems (e.g., nanoparticles, sustained-release formulations) may improve pharmacokinetics

Monitoring Parameters:

  • T cell subset quantification in blood and relevant tissues

  • Bcl-2 expression as a biomarker of IL-7 activity

  • STAT5 phosphorylation for immediate signaling assessment

  • Functional readouts relevant to the disease model

  • Potential increases in viral load in infection models

In SIV infection studies, recombinant glycosylated simian IL-7 administered at 50 μg/kg subcutaneously once weekly for 7 weeks effectively protected CD4+ T cells without significant increases in viral replication, except at the earliest timepoint (day 4) . These parameters may serve as a starting point for rat studies, with appropriate adjustments for species differences.

How effective is IL-7 receptor blockade in rat models of autoimmunity, and what mechanisms underlie these effects?

IL-7 receptor blockade has shown remarkable efficacy in autoimmune disease models, though most published data comes from mouse models with implications for rat studies:

Efficacy in Autoimmune Models:
In nonobese diabetic (NOD) mice, an established model of autoimmune diabetes:

  • Blocking IL-7Rα with monoclonal antibodies prevented autoimmune diabetes

  • When administered to prediabetic mice (10 weeks old, with established islet infiltration), anti-IL-7Rα antibodies reduced diabetes incidence to 10% compared to 60-70% in controls

  • Protection was accompanied by diminished islet infiltration

  • Most remarkably, in new-onset diabetic NOD mice, anti-IL-7Rα treatment restored normoglycemia in approximately 50% of treated animals

Underlying Mechanisms:
Several mechanisms appear to contribute to the therapeutic efficacy:

  • Interference with pathogenic T cell survival and function

  • Potential alteration of T cell trafficking to target tissues

  • Changes in the balance between effector and regulatory T cells

  • Decreased activation of autoreactive T cells

Importantly, IL-7Rα blockade does not appear to selectively deplete islet-specific T effector/memory cells, suggesting that the therapeutic effect is not simply due to elimination of autoreactive cells. Rather, IL-7Rα blockade likely alters the functional properties of these cells .

For researchers conducting similar studies in rat autoimmune models, careful consideration should be given to:

  • The timing of intervention relative to disease onset

  • Antibody specificity and isotype (using antibodies that block without depleting)

  • Comprehensive assessment of various T cell populations and their functional states

  • Long-term follow-up to determine duration of therapeutic effects

What is the potential of IL-7 therapy in rat infection models, and how does this compare to primate studies?

IL-7 therapy shows significant promise in infection models, with evidence primarily from primate studies that can inform rat experimental designs:

Findings from SIV Infection Models:

  • Recombinant, fully glycosylated simian IL-7 (50 μg/kg, s.c., once weekly for 7 weeks) administered throughout the acute phase of SIV infection demonstrated several beneficial effects:

    • Protection from the dramatic decline of circulating naïve and memory CD4+ T cells typically seen in untreated animals

    • Only transient T-cell proliferation in response to IL-7

    • Sustained increase in anti-apoptotic Bcl-2 expression on both CD4+ and CD8+ T cells

    • Persistent expansion of all circulating CD8+ T-cell subsets

    • Development of earlier and stronger SIV Tat-specific T-cell responses

    • Minimal impact on viral replication except at the earliest timepoint

Implications for Rat Infection Models:
When designing rat infection studies, researchers should consider:

  • Selecting appropriate viral or bacterial infection models that induce T cell depletion

  • Optimizing IL-7 dosing based on pharmacokinetic studies in rats

  • Initiating treatment at different disease stages to determine optimal timing

  • Assessing both T cell quantitative and qualitative parameters

  • Monitoring pathogen burden throughout the intervention

  • Evaluating persistence of effects after treatment discontinuation

A significant limitation observed in primate studies was that the beneficial effects of IL-7 were not sustained after treatment interruption . This suggests that continuous or intermittent dosing strategies may be necessary for durable therapeutic effects, which should be explored in rat models.

How should researchers analyze seemingly contradictory data on IL-7 signaling in different rat T cell subsets?

Contradictory findings regarding IL-7 effects on T cell subsets are common in the literature. Resolving these contradictions requires systematic approaches:

Analytical Framework:

  • Verify cell population definitions:

    • Use comprehensive marker panels to precisely identify T cell subsets

    • Distinguish RTEs from mature naïve cells (typically using markers like CD24 or Qa2)

    • Separate CD4+ from CD8+ populations, which may respond differently

    • Consider activation/memory status (using markers like CD44, CD62L)

  • Examine experimental conditions:

    • Standardize IL-7 concentrations across experiments

    • Account for in vitro versus in vivo differences

    • Consider source and glycosylation status of recombinant IL-7

    • Evaluate timing of measurements (some responses may be transient)

  • Employ multiple readouts:

    • STAT5 phosphorylation for immediate signaling

    • Bcl-2 expression for survival effects

    • Proliferation markers for cell division

    • Functional assays for T cell activity

A key example from the literature is the apparent contradiction that RTEs show more efficient IL-7 signaling (higher STAT5 phosphorylation) despite lower IL-7Rα expression and reduced proliferative responses compared to mature naïve T cells. This was resolved by discovering that RTEs preferentially channel IL-7 signals toward Bcl-2 upregulation and survival rather than proliferation .

When facing contradictory data, researchers should:

  • Report all experimental conditions in detail

  • Consider developmental, contextual, and microenvironmental factors

  • Explore mechanistic explanations for divergent findings

  • Use multiple complementary techniques to validate observations

What statistical approaches are most appropriate for analyzing dose-dependent IL-7 effects in heterogeneous T cell populations?

Analyzing IL-7 effects across heterogeneous T cell populations requires sophisticated statistical approaches:

Recommended Statistical Methods:

  • Multivariate analysis:

    • Principal Component Analysis (PCA) to identify patterns across multiple parameters

    • Cluster analysis to identify responding vs. non-responding populations

    • Discriminant analysis to determine which parameters best distinguish treatment effects

  • Mixed-effects models:

    • Account for within-subject correlations in longitudinal studies

    • Handle missing data more effectively than repeated measures ANOVA

    • Incorporate both fixed effects (treatment, dose) and random effects (individual variation)

  • Dose-response modeling:

    • Establish EC50/ED50 values for different cell populations

    • Compare Hill slopes to assess cooperativity of response

    • Use Akaike Information Criterion (AIC) to select optimal models

  • Addressing heterogeneity:

    • Consider stratified analysis for clearly defined subpopulations

    • Employ robust statistical methods less sensitive to outliers

    • Use permutation tests when parametric assumptions are violated

Sample Size Considerations:

  • Power analysis should account for expected variability in T cell responses

  • Higher heterogeneity requires larger sample sizes

  • Consider adaptive designs that allow sample size re-estimation

When reporting results, researchers should:

  • Present both absolute and relative changes in cell populations

  • Provide measures of effect size alongside p-values

  • Include confidence intervals to indicate precision of estimates

  • Distinguish between statistical and biological significance

How do findings from rat IL-7 studies translate to human clinical applications?

Translating IL-7 research from rat models to human applications requires careful consideration of species similarities and differences:

Cross-Species Comparison:

ParameterRatHumanImplications
IL-7 sequence homology~80% with humanReferenceRecombinant human IL-7 may have reduced activity in rats
IL-7Rα distributionSimilar to humansReferenceT cell subset targeting likely translatable
Half-life of IL-7Shorter than in humansLongerDosing frequency may need adjustment
Immune architectureSome differencesReferenceTissue-specific effects may vary

Translational Considerations:

  • Pharmacokinetic bridging:

    • Allometric scaling of doses from rat to human

    • Adjustment for species-specific metabolism

    • Consideration of different administration routes

  • Biomarker validation:

    • Identify conserved biomarkers of IL-7 activity (e.g., STAT5 phosphorylation, Bcl-2 upregulation)

    • Validate surrogate endpoints that predict clinical benefit

    • Develop minimally invasive monitoring approaches

  • Disease model relevance:

    • Assess how well the rat model recapitulates human pathophysiology

    • Consider known species differences in disease progression

    • Evaluate whether therapeutic targets are conserved between species

What are the key considerations for designing rat preclinical studies that most effectively inform human IL-7 clinical trials?

Designing rat preclinical studies with maximum translational value requires attention to several critical factors:

Design Principles for Translational Studies:

  • Mimic clinical scenarios:

    • Study IL-7 effects in clinically relevant disease models

    • Include comorbid conditions common in the target patient population

    • Consider age-appropriate animals when modeling age-related immunodeficiencies

  • Use clinically relevant endpoints:

    • Focus on functional outcomes rather than just mechanistic biomarkers

    • Include quality-of-life measures where possible

    • Assess long-term outcomes, not just acute responses

  • Employ rigorous study design:

    • Randomization and blinding to minimize bias

    • Sample size determination based on power calculations

    • Pre-specified primary and secondary endpoints

    • Inclusion of appropriate control groups

  • Model pharmacological realities:

    • Test clinically feasible dosing regimens

    • Evaluate multiple routes of administration

    • Assess combination therapies with standard-of-care treatments

    • Determine minimal effective dose and therapeutic window

  • Address safety concerns proactively:

    • Monitor for potential enhancement of autoimmunity

    • Assess impact on regulatory T cell populations

    • Evaluate effects on pathogen burden in infection models

    • Screen for off-target effects on non-lymphoid tissues

What are the most promising approaches for enhancing IL-7 efficacy while minimizing potential side effects in rat models?

Several innovative approaches show promise for improving IL-7 therapeutics:

Advanced Delivery Strategies:

  • IL-7/anti-IL-7 antibody complexes for extended half-life

  • Targeted delivery to specific tissues or cell populations

  • Controlled-release formulations for sustained activity

  • Cell-type specific genetic engineering approaches

Molecular Engineering:

  • Optimization of glycosylation patterns for enhanced stability

  • Creation of IL-7 mimetics with selective activity profiles

  • Design of IL-7 fusion proteins with enhanced pharmacokinetics

  • Development of bispecific molecules targeting IL-7R and a second receptor

Combination Approaches:

  • IL-7 with checkpoint inhibitors in cancer immunotherapy

  • IL-7 with antiretrovirals in HIV/SIV models

  • IL-7 with targeted immunosuppressants in transplantation

  • Sequential cytokine therapy regimens (e.g., IL-7 followed by IL-2)

Precision Administration:

  • Biomarker-guided timing of IL-7 administration

  • Individualized dosing based on IL-7 receptor expression

  • Pulsatile rather than continuous administration

  • Tissue-localized delivery for reducing systemic effects

The efficacy and safety of these approaches should be systematically evaluated in rat models before advancing to higher species, with careful attention to both intended immunological effects and potential unwanted consequences .

What emerging technologies will advance our understanding of IL-7 biology in rat T cell development and function?

Cutting-edge technologies are transforming our ability to study IL-7 biology:

Single-Cell Technologies:

  • Single-cell RNA sequencing to identify responder populations

  • CyTOF/mass cytometry for high-dimensional phenotyping

  • Cellular indexing of transcriptomes and epitopes (CITE-seq)

  • Single-cell western blotting for protein analysis

Advanced Imaging:

  • Intravital microscopy to monitor T cell responses in vivo

  • Two-photon imaging of T cell interactions in lymphoid tissues

  • PET imaging with radiolabeled IL-7 to track distribution

  • Histocytometry for spatial analysis of IL-7 responses

Genetic Engineering:

  • CRISPR/Cas9 for precise modification of IL-7 signaling components

  • Conditional knockout models to study cell-type specific responses

  • Reporter rat strains for real-time monitoring of IL-7 signaling

  • Humanized rat models expressing human IL-7R components

Computational Approaches:

  • Machine learning for identifying response patterns

  • Systems biology modeling of IL-7 signaling networks

  • Quantitative pharmacology for dose optimization

  • In silico prediction of IL-7 variant properties

These technologies will enable unprecedented insights into IL-7 biology, including:

  • Identification of novel T cell subsets with distinct IL-7 responsiveness

  • Spatial and temporal dynamics of IL-7 signaling in vivo

  • Integration of IL-7 signals with other cytokine and antigen receptor pathways

  • Prediction of optimal therapeutic strategies for various disease contexts

By leveraging these technologies in rat models, researchers can generate mechanistic insights with greater translational relevance than previously possible .

Product Science Overview

Introduction

Interleukin-7 (IL-7) is a crucial cytokine involved in the regulation of immune responses. It plays a significant role in the development and homeostasis of T cells, B cells, and natural killer (NK) cells. Recombinant IL-7, including rat recombinant IL-7, is widely used in research to study its effects on various immune cells and its potential therapeutic applications.

Discovery and Structure

IL-7 was first discovered in the late 20th century for its growth-promoting effects on B cell progenitors . It is a 25-kDa soluble globular protein produced by various cells, including stromal cells in the bone marrow, thymus, keratinocytes, and enterocytes . The IL-7 receptor (IL-7R) is a heterodimeric complex consisting of the α-chain (CD127) and the common cytokine receptor γ-chain (CD132), which is shared with receptors for other cytokines such as IL-2, IL-4, IL-9, IL-15, and IL-21 .

Biological Functions

IL-7 is essential for the development and survival of T cells in the thymus and the maintenance of peripheral T cells . It also plays a role in B cell maturation and the proliferation and differentiation of memory and naive T cells . IL-7 exerts its biological effects through the activation of the IL-7R, which triggers downstream signaling pathways, including the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway and the phosphatidylinositol 3-kinase (PI3K)-protein kinase B (AKT) pathway .

Recombinant IL-7

Recombinant IL-7, including rat recombinant IL-7, is produced using recombinant DNA technology. It is used in various research applications to study its effects on immune cells and its potential therapeutic uses. Clinical trials of recombinant IL-7 have demonstrated its safety and potent immune reconstitution effects . Recombinant IL-7 has been explored for its potential in cancer immunotherapy, as it can enhance the proliferation and survival of T cells, which are crucial for anti-tumor responses .

Therapeutic Applications

IL-7 has shown promise in various therapeutic applications, particularly in the field of cancer immunotherapy. It has been used to enhance the immune response in patients with cancer by promoting the proliferation and survival of T cells . Additionally, IL-7 has been investigated for its potential to improve immune reconstitution in patients undergoing bone marrow transplantation and those with HIV/AIDS .

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.