IL 32A Human, His

Interleukin-32 alpha Human Recombinant , His Tag
Shipped with Ice Packs
In Stock

Description

Overview of IL-32A Human, His

IL-32A Human, His (Interleukin-32 alpha isoform with a polyhistidine tag) is a recombinant protein produced in Escherichia coli (E. coli). It is a non-glycosylated polypeptide chain comprising 168 amino acids, including residues 1–131 of the native IL-32α isoform fused to a 37-amino acid His-tag at the N-terminus . With a molecular weight of 19.1 kDa, this protein is engineered for enhanced purification and functional studies . IL-32α is one of nine splice variants of the IL-32 gene, located on chromosome 16p13.3, and exhibits distinct biological properties compared to other isoforms .

Key Features:

  • Isoform Specificity: IL-32α lacks 57 amino acids from the C-terminal region present in the longer IL-32γ isoform, contributing to reduced proinflammatory potency .

  • Post-Translational Modification: Susceptible to cleavage by neutrophil-derived Proteinase 3 (PR3) at Thr57-Val58, which enhances its chemokine-inducing activity .

  • Epitope Tags: The His-tag facilitates affinity chromatography purification while retaining bioactivity .

Comparative Activity:

PropertyIL-32αIL-32γ (Reference)
Proinflammatory ActivityModerate High
Antiviral ActivityLow High
Isoform Length131 aa + His-tag231 aa

Production and Purification

IL-32A Human, His is synthesized in E. coli using recombinant DNA technology. The purification process involves:

  1. Affinity Chromatography: Leveraging the His-tag for nickel-based column binding .

  2. Buffer Composition: Stabilized in 20 mM Tris-HCl (pH 8.0), 1 mM DTT, and 10% glycerol for storage .
    This method ensures >95% purity, verified by SDS-PAGE and mass spectrometry .

Immune Modulation:

  • Proinflammatory Signaling: IL-32α induces TNF-α, IL-6, and IL-8 in monocytes and macrophages via NF-κB and p38 MAPK pathways, albeit weaker than IL-32β/γ .

  • Anti-Inflammatory Potential: In HIV-infected individuals, lower IL-32α levels correlate with disease progression, suggesting regulatory roles in chronic inflammation .

Disease Associations:

  • HIV: Plasma IL-32α levels are significantly reduced in typical progressors (TPs) compared to elite controllers (ECs), implicating it in viral control .

  • Cancer: Exhibits paradoxical roles—promotes tumor growth in hepatocellular carcinoma but suppresses it in myeloid leukemias via TNF-α modulation .

Research Applications

IL-32A Human, His is widely used to:

  1. Study isoform-specific cytokine networks in autoimmune diseases (e.g., rheumatoid arthritis) .

  2. Investigate intracellular signaling pathways, including caspase-1-dependent IL-1β maturation .

  3. Develop therapeutic antibodies or inhibitors targeting IL-32-driven inflammation .

Biomarker Potential:

  • IL-32α serves as a predictive biomarker for HIV progression, with lower levels indicating loss of virological control .

  • Total IL-32 (including α/β/γ isoforms) correlates with CD4+ T-cell decline and viral load .

Therapeutic Targets:

  • Inflammatory Diseases: Blocking IL-32α cleavage by PR3 may mitigate neutrophilic inflammation .

  • Cancer: Isoform-specific targeting (e.g., IL-32θ for TNF-α suppression) shows promise in leukemia .

Challenges and Future Directions

  • Isoform-Specific Antibodies: Lack of reagents limits precise detection of IL-32α in complex biological samples .

  • Mechanistic Clarity: Further studies are needed to resolve conflicting roles in cancer and define receptor interactions .

Product Specs

Introduction
IL-32, a member of the cytokine family, plays a critical role in inflammatory responses. It possesses various functional sites, including a tyrosine sulfation site, potential N-myristoylation sites, phosphorylation sites, and an RGD cell-attachment sequence. Elevated IL-32 expression is observed in activated T-cells and NK cells. IL-32 stimulates TNF-a production from macrophages and is involved in the inflammatory response to influenza A virus infection. Dysregulation of IL-32 in myelodysplastic syndrome and chronic myelomonocytic leukemia impacts apoptosis and NK cell function. The p38-MAPK pathway mediates IL-32's induction of TNF, IL-1beta, and IL-6. IL-32 promotes monocyte-to-macrophage differentiation through nonapoptotic, caspase-3-dependent mechanisms. Additionally, IL-32 plays a significant role in the pathogenesis of rheumatoid arthritis and is involved in activation-induced cell death in T cells through intracellular mechanisms. As a cell-associated proinflammatory cytokine, IL-32 is notably stimulated by mycobacteria, leading to the production of interferon gamma via a caspase-1- and IL-18-dependent pathway. Furthermore, IL-32 exhibits associations with TNF-a, IL-1beta, and IL-18, contributing to the development of human rheumatoid arthritis. Targeting IL-32 holds promise for treating autoimmune diseases.
Description
Recombinant human Interleukin-32, expressed in E. coli, is a non-glycosylated polypeptide chain consisting of 168 amino acids (with the IL-32 sequence spanning amino acids 1-131). The protein has a molecular weight of 19.1 kDa. For purification purposes, a 37 amino acid His Tag is fused to the N-terminus of IL-32, enabling purification via conventional chromatography techniques.
Physical Appearance
A clear solution, lacking any color, that has been sterilized through filtration.
Formulation
The IL-32 His Tag protein solution is supplied in a buffer containing 20mM Tris-HCl (pH 8), 1mM DTT, and 10% glycerol.
Stability
For optimal storage, the following guidelines are recommended: - If the entire vial will be used within 2-4 weeks, store at 4°C. - For extended storage, freeze at -20°C. - To further enhance long-term stability, consider adding a carrier protein (0.1% HSA or BSA). - Minimize repeated freeze-thaw cycles to maintain protein integrity.
Purity
The purity of this product exceeds 95.0%, as determined by SDS-PAGE analysis.
Synonyms
NK4, TAIF, TAIFa, TAIFb, TAIFc, TAIFd, IL-32beta, IL-32alpha, IL-32delta, IL-32gamma, Interleukin-32, IL-32, Natural killer cells protein 4, Tumor necrosis factor alpha-inducing factor, IL-32a, IL32a, IL32, Interleukin-32 alpha.
Source
Escherichia Coli.
Amino Acid Sequence
MRGSHHHHHH GMASMTGGQQ MGRDLYDDDD KDRWGSHMCF PKVLSDDMKK LKARMHQAIE RFYDKMQNAE SGRGQVMSSL AELEDDFKEGYLETVAAYYE EQHPELTPLL EKERDGLRCR GNRSPVPDVE DPATEEPGES FCDKSYGAPR GDKEELTPQK CSEPQSSK.

Q&A

What is IL-32A and how does it differ from other IL-32 isoforms?

IL-32A (also known as IL-32α) is one of the four prototypic isoforms of the IL-32 cytokine family, alongside IL-32β, IL-32γ, and IL-32δ. IL-32A is distinguished by its unique structural profile and generally exhibits less pro-inflammatory activity compared to the β and γ isoforms. The human IL-32 gene underwent poor evolutionary conservation, with orthologs showing low homology between species. Notably, IL-32 transcripts are absent in rodents but present in higher mammals including horses, cows, and primates, with primates showing >90% homology to human IL-32 . Understanding these evolutionary differences is crucial when designing animal models for IL-32A research, as traditional rodent models will not express endogenous IL-32 despite responding to the recombinant human protein.

What is the significance of the histidine tag in recombinant IL-32A proteins?

The histidine tag (His-tag) in recombinant IL-32A serves multiple methodological purposes in research applications. This polyhistidine sequence (typically 6-10 histidine residues) facilitates protein purification through immobilized metal affinity chromatography, allowing researchers to isolate the protein with high purity from expression systems. The tag provides a consistent means to detect the protein through anti-His antibodies, which is particularly valuable for tracking IL-32A in experimental systems where specific antibodies might be limited. When working with His-tagged IL-32A, researchers should validate that the tag does not interfere with the protein's biological functions, especially when studying interactions such as the binding between IL-32A and proteinase 3, which has been characterized with a dissociation constant of approximately 2.65×10⁻¹⁰ .

Which cell types express IL-32A, and what stimuli induce its expression?

IL-32 expression spans multiple immune and non-immune cell types, with specific regulation patterns for different isoforms including IL-32A. The table below summarizes key cell types expressing IL-32, the observations regarding its expression, and the stimuli that induce it:

Cell typeObservationsInduced by
MonocytesMediated differentiation of monocytes into dendritic cellsMycobacterium tuberculosis (MTB), Mycobacterium leprae
PBMCsIL-32 synergized with muropeptides for the release of IL-1β and IL-6 in a NOD1- and NOD2-dependent mannerConcanavalin A, Muropeptides, IFN-γ, LPS, HIV, Leishmania, MTB
NK cellsExpressed by NK cell lines and IL-2- and IL-8-activated NK cells; Enhanced NK cell-mediated killing of cancer cells; Expressed in prediagnostic blood samples from children who developed type 1 diabetesIL-2, IL-18, IL-12
T cellsSingle cell sequencing studies revealed Tregs as important sources of IL-32 in various diseases; T-cell lines express IL-32 constitutively; Involved in activation-induced cell deathAnti-CD3 antibody, Ionomycin+PMA, HIV, IL18+IL-12, IL-23, IFN-γ
MacrophagesInduce pro-inflammatory cytokines; IL-32β induces anti-inflammatory IL-10; Induces IDO during HIV infection; Contributes to increased immunity against LeishmaniaTNF-α, IL-15, anti-CD40 antibody, LPS, Poly(I:C), Pam3Cys, MTB, Leishmania, HIV

This expression profile demonstrates the diverse cellular sources of IL-32 and the complex regulatory mechanisms governing its production. Notably, researchers should consider these patterns when designing experiments to study IL-32A specifically, as many stimuli induce multiple isoforms simultaneously.

How do IL-32A levels differ in various disease states, particularly in HIV infection?

IL-32A levels show distinct patterns across different disease states, providing potential biomarker applications. In HIV infection, studies have demonstrated that IL-32A levels vary significantly between different patient groups. HIV-negative individuals and elite controllers (ECs) who naturally control HIV replication showed significantly lower IL-32A levels compared to typical progressors . While IL-32A levels tend to be higher in ECs compared to typical progressors, this difference was not statistically significant in all studies .

When measuring total IL-32 (encompassing all isoforms), typical progressors exhibited significantly higher levels compared to both HIV-negative individuals and elite controllers. This suggests that the more pro-inflammatory isoforms (IL-32β and IL-32γ) rather than IL-32A contribute significantly to the total IL-32 pool in HIV infection . This pattern implies that the balance between different IL-32 isoforms, rather than absolute levels of IL-32A alone, may determine disease outcomes.

Importantly, elevated total IL-32 levels at initial assessment were predictive of subsequent loss of virological control and CD4 T-cell decline in HIV seropositive subjects, demonstrating the potential utility of IL-32 measurement as a prognostic biomarker . Researchers investigating IL-32A specifically should ensure their detection methods can discriminate between isoforms to prevent confounding results.

What are the optimal systems for recombinant IL-32A production and purification?

When producing recombinant IL-32A with a histidine tag, researchers should consider several methodological factors:

Expression Systems: Bacterial systems (particularly E. coli) provide high yield but may not reproduce post-translational modifications. Mammalian expression systems (HEK293 or CHO cells) offer proper folding and modifications but with lower yields. For structural studies requiring high purity, bacterial expression is often preferred, while functional studies may benefit from mammalian-expressed protein.

Purification Protocol: The standard approach involves:

  • Cell lysis under native conditions (unless inclusion bodies form, requiring denaturing conditions)

  • IMAC (Immobilized Metal Affinity Chromatography) using Ni-NTA or Co-NTA resins

  • Gradient elution with imidazole

  • Size exclusion chromatography for final polishing

  • Endotoxin removal for functional studies (critical for cytokine research)

Validation Methods: Verify protein identity and purity through:

  • SDS-PAGE with Coomassie/silver staining (expected MW: ~18-22 kDa plus tag)

  • Western blot with anti-His and anti-IL-32A antibodies

  • Mass spectrometry for precise identification

  • Functional assays comparing activity to commercial standards

Storage Considerations: Store purified IL-32A in buffered solutions (typically PBS with 10% glycerol) at -80°C in single-use aliquots to prevent freeze-thaw degradation. Researchers should validate protein stability through repeated activity testing.

How can researchers accurately differentiate between IL-32 isoforms in experimental samples?

Differentiating between IL-32 isoforms presents significant methodological challenges that researchers must address carefully:

Transcript-Level Discrimination:

  • Design isoform-specific primers spanning unique exon-exon junctions

  • Implement qRT-PCR with validated reference genes

  • Consider digital PCR for absolute quantification

  • RNA-seq followed by isoform-specific bioinformatic analysis

Protein-Level Discrimination:

  • Western blotting with isoform-specific antibodies (when available)

  • Mass spectrometry with peptide mapping to identify unique regions

  • Custom ELISA development using capture/detection antibody pairs with isoform specificity

Challenges and Solutions:

  • Cross-reactivity of antibodies between isoforms requires validation with recombinant standards

  • The lack of specific antibodies for some isoforms (particularly IL-32δ) necessitates indirect detection methods

  • Consider immunoprecipitation followed by mass spectrometry for complex samples

  • When measuring "total IL-32," explicitly document which isoforms the detection method recognizes

Research examining correlations between IL-32 levels and disease states must clearly specify which isoforms are being detected to allow proper interpretation of results.

What is the role of IL-32A in infectious disease immunity?

IL-32A contributes to host defense against pathogens through multiple mechanisms, though its specific role can vary substantially depending on the infectious agent:

Bacterial Infections:
IL-32 production is induced during infections with Mycobacterium tuberculosis (MTB), Helicobacter pylori, and other bacterial pathogens . Various pathogen-associated molecular patterns (PAMPs) upregulate IL-32 expression, including LPS and the double-stranded RNA analog Poly(I:C) . IL-32 appears to function as a "double-edged sword" in bacterial defense - while it mediates potent antimicrobial responses in some contexts, it may promote immune tolerance and support chronic infection in others .

Viral Infections:
IL-32 expression increases during infections with HIV, influenza A, EBV, HPV, and HSV . In HIV infection, conflicting data suggest that IL-32 may both contribute to controlling infection and promote immune suppression . IL-32 was also found to promote latency in EBV infection . The specific contribution of the IL-32A isoform versus other isoforms remains an area requiring further research, as most studies measure total IL-32 or do not discriminate between isoforms.

Methodological Considerations:
When studying IL-32A's role in infectious disease, researchers should:

  • Use isoform-specific detection methods whenever possible

  • Consider both intracellular and secreted IL-32A

  • Examine temporal dynamics of expression throughout infection

  • Assess interactions with other cytokines and immune mediators

  • Validate findings across multiple experimental models

How does IL-32A interact with proteinase 3, and what are the functional implications?

The interaction between IL-32A and proteinase 3 (PR3) represents a crucial regulatory mechanism with significant implications for IL-32 function:

Biochemical Characterization:
PR3 has been identified as a specific binding partner for IL-32A through affinity chromatography experiments using IL-32A-immobilized agarose beads . Surface plasmon resonance analysis determined a dissociation constant of approximately 2.65×10⁻¹⁰, indicating high-affinity binding between urinary or neutrophil-derived PR3 and IL-32A .

Methodological Approaches to Study This Interaction:

  • Pull-down assays: Immobilize recombinant His-tagged IL-32A on Ni-NTA resin and analyze PR3 binding

  • Surface plasmon resonance: Quantify binding kinetics under varying conditions

  • Proximity ligation assays: Visualize interactions in cellular contexts

  • Structural studies: Employ X-ray crystallography or cryo-EM to determine interaction interfaces

Functional Implications:
The IL-32A-PR3 interaction suggests potential regulatory mechanisms where PR3 may:

  • Process IL-32A into more active forms

  • Sequester IL-32A from its downstream targets

  • Modulate IL-32A's ability to induce inflammatory responses

Researchers investigating this interaction should design experiments that can distinguish direct effects of binding from indirect effects on downstream signaling pathways.

How might IL-32A function as an intracellular signaling molecule versus a secreted cytokine?

The dual localization of IL-32A raises fundamental questions about its biological functions:

Intracellular Functions:
Evidence suggests that intracellular IL-32 may promote cell death pathways. Studies using HeLa cells overexpressing IL-32β under tetracycline control demonstrated increased apoptosis . RNA interference reducing IL-32 expression decreased this cell death, supporting the proapoptotic role of intracellular IL-32 . Whether this phenomenon extends to primary cells and specifically to the IL-32A isoform requires further investigation.

Secretion Mechanisms:
The secretion pathway for IL-32 remains incompletely understood. Some reports indicate that T cells activated by PMA and ionomycin or NK cells activated by IL-2 do not actively secrete IL-32, with detected extracellular IL-32 potentially resulting from cell death and leakage . This resembles TNF-α, which functions significantly through its membrane-bound form on T cells despite limited secretion.

Methodological Approaches for Differentiation:
To distinguish intracellular from secreted functions, researchers should:

  • Employ cellular fractionation techniques to locate IL-32A precisely within subcellular compartments

  • Use brefeldin A to block secretion pathways and assess consequences

  • Create non-secretable IL-32A mutants to isolate intracellular functions

  • Develop membrane-impermeable neutralizing antibodies to target only extracellular IL-32A

Future research should investigate whether IL-32A has distinct signaling roles depending on its localization and identify the molecular mechanisms mediating these potentially separate functions.

What is the potential of IL-32A as a biomarker for disease progression, particularly in HIV infection?

Recent research has revealed promising applications for IL-32 isoforms as biomarkers for disease progression:

Evidence from HIV Studies:
Total IL-32 levels were significantly higher in HIV-infected subjects who subsequently lost virological control compared to those who maintained stable viral loads . Importantly, these elevated levels were detectable before the clinical manifestation of disease progression, suggesting predictive value . IL-32 levels positively correlated with CD4 count changes, providing a potential early indicator of immune decline .

The specific contribution of IL-32A versus other isoforms to this biomarker potential requires further clarification, as studies show that IL-32A levels may differ in pattern from total IL-32 levels across disease states .

Methodological Considerations for Biomarker Development:

  • Standardization: Establish consistent quantification protocols using validated reference standards

  • Sampling Considerations: Determine optimal specimen types (plasma, serum, cellular) and processing methods

  • Isoform Specificity: Develop assays that can reliably distinguish IL-32A from other isoforms

  • Clinical Correlation: Conduct longitudinal studies correlating IL-32A levels with clearly defined clinical endpoints

  • Multivariate Analysis: Assess IL-32A in combination with other biomarkers to improve predictive value

Validation Requirements:
For IL-32A to advance as a clinical biomarker, researchers must demonstrate:

  • Analytical validity (precision, accuracy, reproducibility)

  • Clinical validity (sensitivity, specificity, positive/negative predictive values)

  • Clinical utility (impact on patient management decisions)

  • Cost-effectiveness compared to existing prognostic markers

The development of IL-32A as a robust biomarker represents a promising direction requiring concerted efforts to standardize measurement approaches and establish clinical correlations across diverse patient populations.

Product Science Overview

Structure and Production

The human recombinant IL-32 alpha is produced in Escherichia coli (E. coli) as a single, non-glycosylated polypeptide chain. It contains 168 amino acids, with a molecular mass of approximately 19.1 kDa . The recombinant protein is fused to a 37 amino acid His Tag at the N-terminus, which facilitates its purification through conventional chromatography techniques .

Biological Functions

IL-32 alpha is known for its ability to induce the production of other cytokines such as TNF-alpha, IL-1beta, and IL-6 from macrophages . It activates key cytokine signaling pathways, including NF-kappa-B and p38 MAPK . These pathways are crucial for the inflammatory response and play a role in various immune processes.

Role in Diseases

IL-32 alpha has been implicated in several diseases due to its proinflammatory properties. For instance, it is involved in the pathogenesis of rheumatoid arthritis, where it contributes to the chronic inflammation observed in the disease . Additionally, dysregulation of IL-32 has been noted in conditions such as myelodysplastic syndrome and chronic myelomonocytic leukemia, where it modulates apoptosis and impairs NK cell function .

Applications in Research

The recombinant IL-32 alpha with a His Tag is widely used in laboratory research to study its biological functions and role in diseases. It is particularly useful in experiments requiring high purity and specific activity, such as those investigating cytokine signaling pathways and immune responses .

Storage and Stability

For optimal stability, the IL-32 alpha protein should be stored at 4°C if used within 2-4 weeks. For longer-term storage, it is recommended to keep the protein frozen at -20°C, with the addition of a carrier protein like 0.1% HSA or BSA to prevent degradation . It is important to avoid multiple freeze-thaw cycles to maintain the protein’s integrity.

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.