IL17D Human

Interleukin-17D Human Recombinant
Shipped with Ice Packs
In Stock

Description

Interleukin-17D Human Recombinant (18-202) produced in E.Coli is a non-glycosylated disulfide-linked homodimer containing 2 polypeptide chains of 185 amino acids each and having a molecular mass of 40kDa.
The IL-17D is purified by proprietary chromatographic techniques.

Product Specs

Introduction
Interleukin 17D (IL-17D) is a cytokine that plays a role in regulating immune responses. It stimulates the production of molecules involved in inflammation and immunity, such as myeloid growth factors and chemokines. IL-17D also has a role in suppressing the growth of myeloid progenitor cells, which are cells that can develop into various types of blood cells. IL-17D is found in various tissues, including skeletal muscle, heart, adipose tissue, lung, pancreas, and nervous system. It shares structural similarities with IL-17B, another member of the IL-17 family. When IL-17D interacts with endothelial cells, it triggers the production of other signaling molecules like IL-6, IL-8, and CSF2/GM-CSF, contributing to the immune response. This increased production of IL-8 is regulated by a protein complex called NF-kappa B.
Description
This product is a recombinant human IL-17D protein produced in E. coli bacteria. It is a non-glycosylated, disulfide-linked homodimer, meaning it is made up of two identical protein chains, each containing 185 amino acids. The total molecular weight of the protein is 40 kDa. The protein has been purified using specific chromatographic methods to ensure its purity.
Physical Appearance
White powder, freeze-dried and sterilized by filtration.
Formulation
The protein was freeze-dried from a solution containing acetonitrile and TFA that had been passed through a 0.2 micrometer filter.
Solubility
To reconstitute the freeze-dried IL-17D, it is recommended to dissolve it in sterile 18 megaohm-cm water to a concentration of at least 100 micrograms per milliliter. This solution can then be further diluted in other aqueous solutions as needed.
Stability
The lyophilized IL-17D can be stored at room temperature for up to 3 weeks; however, it is recommended to store it in a dry environment below -18 degrees Celsius for extended periods. Once reconstituted, the IL-17D should be stored at 4 degrees Celsius for up to 7 days. For long-term storage, it is advisable to add a carrier protein like HSA or BSA (0.1%) and store it below -18 degrees Celsius. Avoid repeated freeze-thaw cycles.
Purity
The purity of the protein is greater than 97% as determined by SDS-PAGE analysis.
Biological Activity
The recombinant human IL-17D protein exhibits biological activity by binding to the recombinant human IL-17BR protein with an EC50 (half maximal effective concentration) of less than 2 micrograms per milliliter in a binding assay.
Synonyms
Interleukin 17D, Interleukin 27, IL-17D, IL-27, IL27, Interleukin-17D, Interleukin-27, IL17D.
Source
Escherichia Coli.
Amino Acid Sequence
APRAGRRPAR PRGCADRPEE LLEQLYGRLA AGVLSAFHHT LQLGPREQAR NASCPAGGRP ADRRFRPPTN LRSVSPWAYR ISYDPARYPR YLPEAYCLCR GCLTGLFGEE DVRFRSAPVY MPTVVLRRTP ACAGGRSVYT EAYVTIPVGC TCVPEPEKDA DSINSSIDKQ GAKLLLGPND APAGP.

Q&A

What is IL-17D and how does it differ from other IL-17 family members?

IL-17D belongs to the interleukin-17 family, which comprises six members (IL-17A through IL-17F). Unlike other IL-17 family members that generally promote protective immunity and inflammatory responses, IL-17D has immunoregulatory properties that can suppress certain immune responses . While IL-17A and IL-17F are primarily produced by T cells and innate lymphoid cells, IL-17D is predominantly expressed by non-hematopoietic cells, including hepatocytes in steady state .

Methodologically, researchers can distinguish IL-17D from other family members through:

  • Sequence homology analysis showing distinct evolutionary patterns

  • Expression profiling across different tissues and cell types

  • Functional assays demonstrating its unique immunosuppressive effects on dendritic cells (DCs) and CD8 T cells

  • Receptor binding studies examining specific interaction patterns

The distinct functions of IL-17D highlight the importance of studying each cytokine individually rather than extrapolating functions based on family relationships.

What cell types express IL-17D in humans?

IL-17D is primarily expressed by non-hematopoietic cells. Based on research in mouse models, hepatocytes have been identified as significant producers of IL-17D in steady state . The CD45-negative fraction of liver cells expresses higher levels of IL-17D than the CD45-positive fraction, confirming its non-hematopoietic origin .

To investigate IL-17D expression in different human cell types, researchers can employ:

  • qRT-PCR analysis of isolated primary cell populations

  • Immunohistochemistry or in situ hybridization of human tissue samples

  • Single-cell RNA sequencing to identify specific cell populations expressing IL-17D

  • Flow cytometry with intracellular cytokine staining (if suitable antibodies are available)

  • Cell sorting followed by transcriptional analysis

Interestingly, IL-17D expression is dynamically regulated during infection, with evidence of decreased expression in hepatocytes during Listeria monocytogenes infection . Both mRNA and protein levels are reduced after infection, suggesting that understanding the context-dependent regulation of IL-17D expression is critical for elucidating its function.

What are the standard methods for detecting IL-17D in human samples?

Several methodological approaches can be used to detect and quantify IL-17D in human samples:

  • ELISA (Enzyme-Linked Immunosorbent Assay):

    • Useful for quantifying IL-17D protein in serum, plasma, or tissue lysates

    • Has been successfully used to measure IL-17D protein in liver tissue before and after infection

    • Optimal for high-throughput screening but may have sensitivity limitations

  • qRT-PCR (Quantitative Reverse Transcription PCR):

    • Enables measurement of IL-17D mRNA expression levels

    • Highly sensitive for detecting transcriptional changes

    • Requires careful primer design to distinguish IL-17D from other family members

    • Has been used to demonstrate dynamic regulation of IL-17D in hepatocytes

  • Western Blotting:

    • Allows detection of IL-17D protein and assessment of molecular weight

    • Requires validated antibodies with confirmed specificity

    • Useful for confirming protein expression in cell or tissue lysates

  • Flow Cytometry:

    • Useful for intracellular detection of IL-17D in specific cell populations

    • Requires permeabilization and validated antibodies

    • Can be combined with surface markers to identify producing cell types

Researchers should consider the limitations of each method, including sensitivity, specificity, and the distinction between mRNA and protein detection. Cross-validation using multiple techniques is recommended for robust results.

How is IL-17D expression regulated under different physiological conditions?

The regulation of IL-17D expression appears to be context-dependent and differs from other IL-17 family members. Based on experimental evidence:

  • Baseline Expression:

    • Constitutively expressed in non-hematopoietic cells, particularly hepatocytes in steady state

    • Expression levels vary across different tissues

  • Infection-Induced Regulation:

    • IL-17D expression in hepatocytes progressively decreases during Listeria monocytogenes infection

    • Both mRNA and protein levels are reduced following infection

    • This downregulation may represent a mechanism to enhance protective immunity

  • Potential Regulatory Mechanisms:

    • Transcriptional regulation by various transcription factors

    • Post-transcriptional regulation through mRNA stability

    • Potential feedback mechanisms from inflammatory mediators

To study IL-17D regulation, researchers can employ:

  • Reporter gene constructs to identify promoter elements

  • ChIP-seq to identify transcription factor binding sites

  • Time-course experiments during different physiological challenges

  • Pharmacological inhibitors to dissect signaling pathways involved in regulation

Understanding the regulatory mechanisms of IL-17D expression is crucial for developing potential therapeutic approaches targeting this cytokine in different disease contexts.

What is the role of IL-17D in regulating CD8 T cell responses during infection?

IL-17D plays a unique immunoregulatory role in suppressing CD8 T cell responses during infection. This function distinguishes it from other IL-17 family members that typically enhance protective immunity.

Research findings indicate that:

  • IL-17D-deficient mice exhibit enhanced antigen-specific CD8 T cell responses during Listeria monocytogenes infection

  • The enhanced CD8 T cell activity correlates with reduced pathogen burden in IL-17D-deficient mice

  • This protective phenotype is observed in both bacterial (Listeria) and viral (influenza A) infections

  • The suppressive effect of IL-17D on CD8 T cells appears to be indirect, mediated through dendritic cells

Methodological approaches to investigate IL-17D's role in CD8 T cell regulation include:

  • In vivo models:

    • Comparison of antigen-specific CD8 T cell responses in wild-type versus IL-17D-deficient mice

    • Adoptive transfer experiments to track antigen-specific T cell proliferation and function

    • Temporal analysis of T cell responses at different stages of infection

  • Ex vivo systems:

    • Co-culture of dendritic cells with CD8 T cells in the presence or absence of recombinant IL-17D

    • Assessment of T cell proliferation using dye dilution assays (e.g., CellTrace Violet)

    • Measurement of effector functions (cytokine production, cytotoxicity)

These findings suggest that IL-17D represents a regulatory mechanism that may prevent excessive CD8 T cell responses during infection. Understanding this pathway could provide insights into immune homeostasis and potential therapeutic targets for enhancing anti-pathogen immunity.

How does IL-17D modulate dendritic cell function and subsequent T cell activation?

IL-17D directly impacts dendritic cell (DC) function, which subsequently affects T cell activation. Research using mouse models and ex vivo systems has revealed:

  • Direct Effects on Dendritic Cells:

    • IL-17D suppresses the expression of costimulatory molecules (e.g., B7.1/CD80) on DCs

    • IL-17D treatment reduces cytokine production (IL-6 and IL-12) by DCs

    • These effects were observed in DCs isolated from Listeria-infected mice and treated ex vivo with IL-17D

  • Consequences for T Cell Activation:

    • In DC/CD8 T cell co-culture systems, IL-17D suppresses:

      • Proliferation of antigen-specific CD8 T cells (OT-1 TCR transgenic cells)

      • IFNγ production by CD8 T cells

    • These effects are concentration-dependent (tested at 0-200 ng/ml)

    • Heat-inactivated IL-17D does not affect cytokine production, indicating the specificity of the effect

  • In Vivo Relevance:

    • In RAG-deficient mice (lacking adaptive immunity), IL-17D-deficient animals show:

      • Increased number of DCs

      • Enhanced activation status of DCs after Listeria infection

    • This suggests that IL-17D regulates DC activation independent of adaptive immune cells

Methodological approaches to study IL-17D's effects on DC function include:

  • Isolation of DCs from infected mice for ex vivo studies

  • Flow cytometric analysis of DC activation markers (CD80, CD86, and MHC I)

  • Cytokine profiling of DC culture supernatants

  • DC/T cell co-culture systems with titrated amounts of recombinant IL-17D

These findings reveal a novel immunoregulatory pathway in which IL-17D from non-hematopoietic sources suppresses DC activation, subsequently limiting CD8 T cell responses during infection.

What are the conflicting findings regarding IL-17D's role in host defense against different pathogens?

Research on IL-17D has yielded seemingly contradictory findings regarding its role in host defense:

  • Findings Supporting Immunosuppressive Role:

    • IL-17D-deficient mice showed resistance to Listeria monocytogenes infection with:

      • Reduced bacterial burden in liver and spleen

      • Enhanced antigen-specific CD8 T cell responses

      • Improved survival rates (50% survival vs. 0% in wild-type)

    • Similar protective phenotype observed in IL-17D-deficient mice during influenza A virus infection

  • Contradictory Findings in Other Infection Models:

    • Previous reports described enhanced infection in IL-17D-deficient mice infected with:

      • Vaccinia virus (administered via scarification)

      • Murine cytomegalovirus (administered intraperitoneally)

    • These findings suggest IL-17D may have protective functions in certain infection contexts

  • Potential Factors Explaining Discrepancies:

    • Pathogen type (bacterial vs. different viral pathogens)

    • Route of infection (intravenous vs. scarification vs. intraperitoneal)

    • Timing of analysis (acute vs. chronic phases)

    • Tissue-specific effects

Methodological approaches to resolve these conflicts:

  • Comparative studies using standardized infection protocols

  • Temporal analysis throughout infection course

  • Tissue-specific conditional knockout models

  • Cross-pathogen studies in the same experimental system

What experimental models are most suitable for studying IL-17D function in human systems?

Given the complexities of studying IL-17D in human systems, researchers can employ several complementary experimental models:

  • Human Primary Cell Systems:

    • Isolation and culture of primary human hepatocytes (potential IL-17D producers)

    • Human dendritic cell cultures treated with recombinant IL-17D

    • Co-culture systems with human DCs and T cells (similar to mouse models)

    • Organ-specific cell cultures (depending on research question)

  • Human Cell Lines:

    • Hepatocyte cell lines for IL-17D expression studies

    • Reporter cell lines engineered to express IL-17D receptors

    • CRISPR/Cas9-modified cell lines with IL-17D knockout or overexpression

  • Ex Vivo Human Tissue Models:

    • Precision-cut liver slices for studying IL-17D expression and regulation

    • Human tissue explant cultures from surgical specimens

    • Organoid cultures representing different human tissues

  • Humanized Mouse Models:

    • Mice reconstituted with human immune system components

    • Human tissue xenografts in immunodeficient mice

    • Transgenic mice expressing human IL-17D or IL-17D receptors

  • Translational Approaches:

    • Analysis of clinical samples from patients with infections

    • Correlation of IL-17D levels with disease parameters

    • Single-cell RNA sequencing of human tissue samples

Methodological considerations for human IL-17D research:

  • Validation of antibody specificity for human IL-17D detection

  • Optimization of culture conditions for primary human cells

  • Careful selection of recombinant protein sources and quality control

  • Technical challenges in detecting low-abundance cytokines

Combining multiple approaches provides the most robust understanding of IL-17D function in human biology.

What are the methodological considerations when designing experiments to study IL-17D signaling pathways?

Designing rigorous experiments to elucidate IL-17D signaling pathways requires careful methodological considerations:

  • Receptor Identification and Validation:

    • Determine which receptor components mediate IL-17D signaling

    • Validate receptor expression in target cells using:

      • qRT-PCR for transcript detection

      • Flow cytometry or western blotting for protein expression

      • siRNA/shRNA knockdown to confirm receptor specificity

  • Recombinant Protein Quality:

    • Source high-quality recombinant IL-17D with:

      • Confirmed bioactivity

      • Low endotoxin levels

      • Proper folding and post-translational modifications

    • Include appropriate controls:

      • Heat-inactivated protein (as used in published studies)

      • Isotype control proteins

      • Dose-response curves (e.g., 0-200 ng/ml as used in studies)

  • Signaling Pathway Analysis:

    • Temporal analysis of signaling events (minutes to hours)

    • Phosphorylation status of potential downstream mediators:

      • MAP kinases (ERK, p38, JNK)

      • NF-κB pathway components

      • STAT proteins

    • Use of pharmacological inhibitors with appropriate controls

    • Genetic approaches (siRNA, CRISPR) to validate pathway components

  • Functional Readouts:

    • Cell type-specific functional assays:

      • For DCs: surface marker expression (CD80, CD86, MHC I), cytokine production (IL-6, IL-12), T cell stimulatory capacity

      • For T cells: proliferation (measured by dye dilution), cytokine production (IFNγ)

    • Multiplex cytokine analysis of culture supernatants

    • Phenotypic changes (morphology, proliferation, survival)

  • Data Analysis Considerations:

    • Appropriate statistical tests for experimental design

    • Normalization methods for gene expression data

    • Validation of key findings using complementary approaches

By systematically addressing these methodological considerations, researchers can generate robust and reproducible data on IL-17D signaling pathways.

How can contradictory data about IL-17D function be reconciled in experimental design?

Reconciling contradictory findings about IL-17D function requires careful experimental design strategies:

  • Standardized Experimental Systems:

    • Use consistent genetic backgrounds for animal models

    • Implement identical infection protocols across studies

    • Adopt uniform readout parameters and analysis timepoints

  • Context-Dependent Analysis:

    • Systematically vary parameters to identify context-dependent effects:

      • Pathogen type and dose

      • Route of administration (intravenous, intraperitoneal, scarification)

      • Tissue microenvironment

      • Acute versus chronic settings

  • Comprehensive Phenotyping:

    • Analyze multiple cell types and tissues simultaneously

    • Perform temporal analysis throughout infection course

    • Measure both local and systemic responses

    • Integrate cellular, molecular, and functional readouts

  • Genetic Approaches:

    • Use conditional knockout models to dissect cell type-specific contributions

    • Generate bone marrow chimeras to determine contribution of hematopoietic vs. non-hematopoietic compartments

    • Cross IL-17D-deficient mice with other relevant knockout strains

  • Mechanistic Validation:

    • Confirm phenotypes with multiple independent approaches

    • Validate key findings using gain-of-function and loss-of-function strategies

    • Perform rescue experiments with recombinant protein administration

A specific experimental design to reconcile contradictory findings might include:

  • Parallel infection of IL-17D-deficient and control mice with multiple pathogens

  • Multiple routes of administration for each pathogen

  • Comprehensive immune phenotyping at standardized timepoints

  • Analysis of tissue-specific IL-17D expression dynamics

  • Ex vivo functional assays with cells from infected animals

By systematically addressing potential sources of variability, researchers can develop a more nuanced understanding of IL-17D's context-dependent functions.

What are the molecular mechanisms underlying IL-17D-mediated suppression of immune responses?

The molecular mechanisms by which IL-17D suppresses immune responses, particularly DC activation and subsequent CD8 T cell responses, are not fully elucidated. Based on available research, several potential mechanisms can be proposed:

  • Receptor Engagement and Proximal Signaling:

    • Identification of the specific receptor complex for IL-17D on DCs

    • Characterization of immediate signaling events following receptor engagement

    • Potential competitive inhibition with other IL-17 family members

  • Transcriptional Regulation in Dendritic Cells:

    • IL-17D treatment reduces expression of costimulatory molecules (CD80/B7.1)

    • Suppresses production of T cell-activating cytokines (IL-6, IL-12)

    • May involve:

      • Inhibition of positive transcription factors

      • Activation of negative regulators

      • Epigenetic modifications affecting gene accessibility

  • Antigen Processing and Presentation:

    • Effects on MHC class I loading and trafficking

    • Alteration of costimulatory molecule distribution on cell surface

    • Impact on antigen-presenting capacity

Methodological approaches to investigate these mechanisms:

  • Phospho-flow cytometry to assess signaling pathway activation

  • RNA-seq and proteomics of IL-17D-treated DCs

  • CRISPR screening to identify essential components of IL-17D signaling

  • Ex vivo DC/CD8 T cell co-culture systems with specific pathway inhibitors

A detailed understanding of these molecular mechanisms could:

  • Reveal novel immunoregulatory pathways

  • Identify potential therapeutic targets for modulating immunity

  • Explain the seemingly contradictory roles of IL-17D in different infection contexts

  • Provide insights into the evolution of the IL-17 cytokine family and functional diversification

Product Science Overview

Structure and Expression

IL-17D is a secreted cytokine with a conserved cysteine-knot fold near the C-terminus, similar to other IL-17 family members . It is expressed in various tissues, including skeletal muscle, heart, adipose tissue, lung, pancreas, and the nervous system . The recombinant form of IL-17D is typically produced in E. coli and has a predicted molecular mass of approximately 20 kDa per monomer .

Biological Functions

IL-17D modulates immune responses indirectly by stimulating the production of myeloid growth factors and chemokines . It has been shown to stimulate the production of cytokines such as IL-6, IL-8, and GM-CSF in endothelial cells . Additionally, IL-17D has an inhibitory effect on the hemopoiesis of myeloid progenitor cells .

Role in Disease and Therapeutic Potential

The IL-17 family, including IL-17D, has been linked to various disease processes such as rheumatoid arthritis, asthma, lupus, allograft rejection, tumorigenicity, and antitumor immunity . IL-17D, in particular, has been found to be highly secreted by fibrosarcoma tumor cells and can recruit natural killer cells via the production of CCL2 by endothelial cells .

Applications of Recombinant IL-17D

Recombinant IL-17D is used in research to study its role in immune responses and its potential therapeutic applications. It is available in both carrier-free and carrier-containing formulations, with the carrier protein typically being Bovine Serum Albumin (BSA) to enhance protein stability . The recombinant protein is lyophilized and can be reconstituted for use in various experimental setups .

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.