C-JUN Human

Jun Proto-Oncogene Human Recombinant
Shipped with Ice Packs
In Stock

Description

C-JUN amino acids 1-81 produced in E.coli, is a non-glycosylated, polypeptide chain having a molecular mass of 52 kDa.
C-JUN is a maltose binding protein (MBP) fusion protein with an amino-terminal polyhistidine tag and purified by proprietary chromatographic techniques.

Product Specs

Introduction
The C-JUN gene partners with the c-Fos gene to create the AP-1 early response transcription factor. This factor is activated by the JNK pathway. C-JUN is believed to be the transforming gene of avian sarcoma virus 17. The C-JUN protein closely resembles the viral protein and directly interacts with specific DNA sequences to regulate gene expression. Notably, the C-JUN gene lacks introns and resides at the chromosomal location 1p32-p31, a region often implicated in translocations and deletions within human cancers.
Description
This product consists of C-JUN amino acids 1-81 produced in E.coli. It is a non-glycosylated polypeptide chain with a molecular weight of 52 kDa. The C-JUN is fused with a maltose binding protein (MBP) and carries an amino-terminal polyhistidine tag. Purification is achieved using proprietary chromatographic methods.
Physical Appearance
Sterile Filtered White lyophilized (freeze-dried) powder.
Formulation
C-JUN is provided as a lyophilized powder without any additional ingredients.
Solubility
Before opening, it is advisable to centrifuge the vial to ensure the contents are at the bottom. For reconstitution, dissolve the lyophilized c-Jun in 40mM Tris, pH 7.5, aiming for a final concentration between 0.2-1.0 mg/ml.
Stability
For short-term storage (1-2 weeks), the product can be kept at 4°C. For extended periods, store frozen at -20°C. Minimize repeated freezing and thawing.
Biological Activity
C-Jun can be phosphorylated in vitro using either recombinant active JNK1 or JNK2, or JNK immunoprecipitated from stimulated cells. Phosphorylation can be tracked via Western blot analysis using a c-Jun [pS73] specific antibody, combined with chemiluminescence detection. It's important to note that optimizing the cell stimulation protocol, cell lysis procedure, and reaction conditions might be necessary for specific applications.
Note
The observed kinase activity might fluctuate depending on the substrate and the reaction conditions used.
Purity
Purity exceeds 95% as assessed by SDS-PAGE.
Synonyms
Transcription factor AP-1, Activator protein 1, AP1, Proto-oncogene c-jun, V-jun avian sarcoma virus 17 oncogene homolog, p39, c-Jun.
Source
Escherichia Coli.

Q&A

What is c-JUN and what are its primary functions in human cells?

c-JUN is a proto-oncogene and a major component of the activator protein-1 (AP-1) transcription factor complex. It belongs to the basic-Leucine Zipper (bZIP) family of transcription factors . The protein encoded by c-JUN (gene symbol: JUN, Gene ID: 3725) is also known as Transcription factor AP-1, Activator protein 1, Proto-oncogene c-Jun, or p39 .

As a transcription factor, c-JUN regulates gene expression by binding to specific DNA sequences as either homodimers or heterodimers with other AP-1 family members (including Fos, ATF, BATF, and JDP2 proteins) . This versatility in dimerization partners allows c-JUN to integrate diverse extrinsic and intrinsic signals, resulting in context-dependent regulation of gene expression.

c-JUN is involved in multiple cellular processes including:

  • Cell proliferation and cell cycle regulation

  • Cellular differentiation

  • Apoptosis

  • Malignant transformation in cancer development

  • Immune cell function and activation

  • Embryonic development and stem cell differentiation

The multifaceted nature of c-JUN allows it to elicit distinct cellular responses in different cell types and under various stimulation conditions .

How can researchers measure c-JUN transcription factor activity in experimental settings?

Researchers can measure c-JUN transcription factor activity using specialized assays that detect DNA-binding capacity of active c-JUN. A commonly used approach is the Transcription Factor Activity Assay, which provides semi-quantitative assessment of c-JUN activity .

The methodological approach involves:

  • Sample preparation: Researchers prepare cell lysates or nuclear extracts from cells of interest.

  • DNA-binding reaction: The assay uses double-stranded DNA (dsDNA) coated plates with canonical c-JUN binding sequences. Active c-JUN in the samples binds to these sequences.

  • Detection: Bound c-JUN is detected using specific antibodies followed by a colorimetric detection system.

  • Analysis: Results are typically analyzed as relative activity levels compared to controls.

This approach allows researchers to compare c-JUN activity under different experimental conditions, such as after gene knockdown or in response to stimuli like TNF-alpha. For example, researchers at Washington State University have successfully used such assays to test c-JUN DNA binding activity in nuclear extracts from rheumatoid arthritis synovial fibroblasts, comparing activity levels in non-stimulated versus TNF-alpha-stimulated conditions .

For more comprehensive analysis, researchers often combine activity assays with other methods such as Western blotting to measure total c-JUN protein levels, quantitative PCR to assess mRNA expression, and chromatin immunoprecipitation (ChIP) to identify genomic binding sites.

What mechanisms underlie c-JUN's role in cancer development, particularly in glioblastoma?

c-JUN contributes to cancer development through several mechanisms, with translational regulation being particularly significant in glioblastoma. Research has revealed that c-JUN accumulation is robustly elevated in human glioblastoma, contributing directly to the malignant properties of these cells .

The key mechanistic findings include:

  • Translational activation via IRES: The c-JUN 5′UTR harbors a potent internal ribosomal entry site (IRES) with a virus-like domain that directs cap-independent translation in glioblastoma cells. This means c-JUN protein accumulation occurs with no corresponding increase in c-JUN mRNA or extended protein half-life, but rather through enhanced translation efficiency of existing transcripts .

  • Independence from MAPK signaling: Unlike typical c-JUN activation, the increased c-JUN accumulation in glioblastoma is not dependent on MAPK activity but can be stimulated by a cytoskeleton-dependent pathway .

  • Contribution to malignant properties: Elevated c-JUN levels enhance the malignant phenotype of glioblastoma cells, suggesting it plays a direct role in cancer progression.

This translational control mechanism represents a previously undescribed pathway of c-JUN regulation that might be relevant to other types of human cancer. The unique structural and functional properties of the c-JUN IRES element offer potential targets for therapeutic interventions aimed at down-regulating c-JUN in cancer cells .

How does c-JUN regulate mesoderm differentiation in human pluripotent stem cells?

c-JUN functions as a chromatin repressor that limits mesoderm differentiation as cells exit pluripotency. This regulation occurs through specific epigenetic mechanisms .

The experimental evidence demonstrates:

  • c-JUN is widely expressed across various cell types in early embryogenesis but is not essential for maintaining pluripotency. Instead, it functions as a repressor specifically constraining mesoderm development while having minimal impact on ectoderm differentiation .

  • Mechanism of repression: c-JUN interacts with the MBD3-NuRD (Nucleosome Remodeling and Deacetylase) complex, maintaining chromatin in a low accessibility state at mesoderm-related genes during the differentiation of human pluripotent stem cells (hPSCs) .

  • Target gene specificity: c-JUN specifically inhibits the activation of key mesoderm factors, such as EOMES and GATA4, which are essential for mesoderm development .

  • Experimental manipulation outcomes:

    • Knocking out c-JUN or inhibiting it with a JNK inhibitor relieves this suppression, promoting mesoderm cell differentiation

    • Similarly, knockdown of MBD3 enhances mesoderm generation, while MBD3 overexpression impedes it

    • Overexpressing c-JUN redirects differentiation toward a fibroblast-like lineage

These findings reveal that c-JUN acts as a chromatin regulator to restrict mesoderm cell fate during early human embryonic development. The dynamic expression pattern of c-JUN during differentiation is also noteworthy - it is initially downregulated on day 1 but reactivated by day 3, suggesting that c-JUN needs to be repressed during the early stage of hPSC differentiation into mesoderm .

What role does c-JUN play in CD8 T cell function and exhaustion in immune responses?

c-JUN/AP-1 plays a critical role in regulating CD8 T cell responses to both acute infection and cancer, with its activity level affecting the balance between effective immune function and T cell exhaustion .

The key aspects of c-JUN's role in CD8 T cells include:

  • Productive immune response: c-JUN/AP-1 participates in the transcriptional and epigenetic regulatory mechanisms that drive effective CD8 T cell immune responses. High levels of c-JUN activity are associated with effector T cell functions .

  • T cell exhaustion mechanisms: Downregulation of c-JUN contributes to the dysfunctional state of tumor-infiltrating CD8 T cells. Exhausted T cells show reduced c-JUN/AP-1 activity and increased expression of transcription factors like TOX and NR4A, which promote exhaustion .

  • Chromatin landscape changes: CAR T cells deficient in exhaustion-promoting factors (TOX or NR4A) exhibit a gene expression profile characteristic of CD8 effector T cells, with open chromatin regions enriched for both NF-κB and c-JUN/AP-1 binding motifs .

  • Therapeutic potential: CAR T cells engineered to overexpress c-JUN show resistance to exhaustion, suggesting that the JNK/c-JUN pathway is a suitable target for immunotherapy-based approaches to reinvigorate anti-tumor immune functions .

The transcriptional program controlled by c-JUN in CD8 T cells integrates various extracellular signals to determine cell fate decisions between effective anti-tumor responses and exhaustion. This makes c-JUN a promising target for improving immunotherapeutic strategies in cancer treatment .

What genetic manipulation techniques are most effective for studying c-JUN function in human cells?

Several genetic manipulation approaches have proven effective for studying c-JUN function in human cells, with CRISPR/Cas9-based knockout systems being particularly valuable. Based on the research literature, the following methodological approaches have been successful:

  • CRISPR/Cas9 knockout system:

    • Utilize pairs of sgRNAs to delete target exons in c-JUN

    • Specific sgRNA sequences that have proven effective include: sgRNA1: acaagtttcggggccgcaac; sgRNA2: gagaacttgacaagttgcga

    • Selection with puromycin followed by single clone expansion and validation

    • Validation through genomic DNA extraction, PCR analysis, Sanger sequencing, western blotting, and karyotype analysis

  • Inducible overexpression systems:

    • PiggyBac (PB) transposon-based expression systems (e.g., PB-TRE3G-c-JUN-SV40 polyA)

    • Doxycycline-inducible expression for temporal control of c-JUN levels

    • Constitutive expression systems (e.g., PB-CAG-c-JUN) for stable overexpression

  • RNA interference approaches:

    • siRNA transfection for transient knockdown

    • shRNA for stable knockdown

    • Electroporation-based transfection (6 cells seeded on Matrigel-coated wells in appropriate medium supplemented with 10 μM Y27632)

  • Pharmacological inhibition:

    • JNK inhibitors (e.g., SP600125) to block c-JUN activation by preventing phosphorylation

    • Useful for distinguishing between genomic and non-genomic functions of c-JUN

These approaches can be combined with functional assays and genomic analyses (RNA-seq, ATAC-seq, ChIP-seq) to comprehensively understand c-JUN's role in specific cellular contexts and processes.

What are the key considerations for analyzing c-JUN-dependent gene expression changes?

Analyzing c-JUN-dependent gene expression changes requires careful experimental design and consideration of several key factors:

  • Cell type and context specificity:

    • c-JUN functions differently across cell types (e.g., pluripotent stem cells vs. differentiated cells)

    • Context-dependent effects must be considered (e.g., different outcomes in cancer vs. normal cells)

    • Appropriate control cell lines must be established

  • Dimerization partner analysis:

    • c-JUN can form heterodimers with various AP-1 family members

    • Different dimerization partners lead to distinct gene expression outcomes

    • Consider co-immunoprecipitation studies to identify relevant binding partners in your system

  • Temporal dynamics:

    • c-JUN expression and activity change dynamically (e.g., downregulated on day 1 of mesoderm differentiation but reactivated by day 3)

    • Time-course experiments are essential to capture these dynamics

    • Consider inducible systems for precise temporal control

  • Multi-omics integration:

    • Combine RNA-seq with chromatin accessibility data (ATAC-seq)

    • Include ChIP-seq to identify direct c-JUN binding sites

    • Integrate with proteomics data to account for post-transcriptional regulation

  • Bioinformatic analysis approaches:

    • Motif enrichment analysis to identify AP-1 binding sites

    • Pathway analysis to understand biological processes affected

    • Integration with relevant public datasets (e.g., human gastrulation datasets)

    • Single-cell analyses to account for cellular heterogeneity

  • Functional validation:

    • Include rescue experiments to confirm specificity

    • Use multiple genetic manipulation approaches (knockout, knockdown, inhibitors)

    • Validate key findings with independent molecular techniques

When publishing results, researchers should clearly report the specific AP-1 family members studied, the cellular context, temporal aspects, and consider the translational regulation of c-JUN, which may result in discrepancies between mRNA and protein levels .

How do different post-translational modifications affect c-JUN function?

c-JUN undergoes various post-translational modifications that significantly impact its activity, stability, and interaction capabilities. Understanding these modifications is essential for comprehending the multifaceted output of c-JUN biological activity:

  • Phosphorylation:

    • JNK-mediated phosphorylation at Ser63 and Ser73 enhances c-JUN transactivation potential

    • Inhibition of JNK with inhibitors like SP600125 prevents c-JUN activation

    • Different phosphorylation patterns can lead to distinct functional outcomes

    • Phosphorylation status affects protein stability and degradation rates

  • Ubiquitination:

    • Regulates c-JUN protein turnover

    • E3 ubiquitin ligases target c-JUN for proteasomal degradation

    • Phosphorylation status can influence ubiquitination patterns

  • Acetylation:

    • Affects DNA binding and transcriptional activity

    • Can modify interaction with chromatin-modifying complexes like MBD3-NuRD

    • May influence dimerization partner selection

  • SUMOylation:

    • Can repress c-JUN transcriptional activity

    • Affects nuclear localization and protein-protein interactions

The complex interplay between these modifications creates a "PTM code" that determines c-JUN activity in different cellular contexts. For example, in mesoderm differentiation, the interaction between c-JUN and the MBD3-NuRD complex is likely regulated by specific post-translational modification patterns . Similarly, in cancer cells like glioblastoma, altered modification patterns may contribute to abnormal c-JUN activity and protein stability .

Researchers studying c-JUN should consider using phospho-specific antibodies, proteasome inhibitors, and mass spectrometry approaches to comprehensively analyze the PTM status of c-JUN in their experimental systems.

What are the emerging therapeutic strategies targeting c-JUN in cancer and immune-related diseases?

Several emerging therapeutic strategies targeting c-JUN are being developed for cancer and immune-related diseases, based on growing understanding of its role in these pathologies:

  • IRES-targeted approaches for cancer:

    • The discovery that c-JUN mRNA contains an IRES element that drives cap-independent translation in glioblastoma offers a novel therapeutic target

    • Small molecules or oligonucleotides that disrupt the IRES structure could selectively inhibit c-JUN translation in cancer cells

    • This approach could potentially overcome resistance to conventional therapies that target upstream signaling pathways

  • JNK inhibition strategies:

    • JNK inhibitors (e.g., SP600125) can prevent c-JUN activation

    • In stem cell differentiation contexts, JNK inhibitors promote mesoderm differentiation by relieving c-JUN-mediated repression

    • Selective JNK inhibitors with improved specificity are being developed as potential therapeutics

  • Engineered T cells for immunotherapy:

    • CAR T cells engineered to overexpress c-JUN show resistance to exhaustion

    • This approach could improve the efficacy of CAR T cell therapy for solid tumors

    • Combination with checkpoint inhibitors may provide synergistic effects

  • Targeting c-JUN interaction partners:

    • Disrupting the interaction between c-JUN and the MBD3-NuRD complex could be a strategy to modulate stem cell differentiation

    • Small molecules targeting specific protein-protein interactions could provide more selective modulation than direct c-JUN inhibition

  • Epigenetic approaches:

    • Since c-JUN functions as a chromatin regulator, combining c-JUN targeting with epigenetic modulators may enhance therapeutic efficacy

    • HDAC inhibitors or other epigenetic drugs might synergize with c-JUN/JNK targeting approaches

The therapeutic strategy should be tailored to the specific disease context, as c-JUN can function as either an oncogene or tumor suppressor depending on the cellular environment. For example, while inhibiting c-JUN may be beneficial in glioblastoma , enhancing c-JUN activity might be preferred in cases of T cell exhaustion in the tumor microenvironment .

How do contradictory findings about c-JUN function across different cell types reconcile with its evolutionary conservation?

The seemingly contradictory roles of c-JUN across different cell types reflect its evolution as a context-dependent transcriptional regulator that integrates multiple signaling inputs to produce cell type-specific responses. Several factors help reconcile these apparent contradictions:

  • Dimerization partner diversity:

    • c-JUN can form heterodimers with various AP-1 family members (Fos, ATF, BATF, JDP2)

    • Different dimerization partners result in distinct DNA binding specificities and transcriptional outputs

    • The relative abundance of potential partners varies across cell types

  • Cell type-specific co-factor interactions:

    • In stem cells, c-JUN interacts with the MBD3-NuRD complex to repress mesoderm genes

    • In immune cells, c-JUN may interact with lineage-specific factors to regulate T cell function

    • These differential protein-protein interactions enable context-dependent functions

  • Chromatin landscape variations:

    • The accessible chromatin landscape differs between cell types

    • c-JUN binding sites may be accessible in some cell types but not others

    • Pre-existing epigenetic marks influence whether c-JUN binding results in activation or repression

  • Integration with other signaling pathways:

    • c-JUN acts as an integration point for multiple signaling pathways

    • The specific combination of active pathways in different cell types determines the outcome of c-JUN activity

    • For example, in glioblastoma, c-JUN translation is regulated by a cytoskeleton-dependent pathway rather than the typical MAPK pathway

  • Temporal dynamics of expression and activity:

    • c-JUN expression and activity are dynamically regulated

    • During mesoderm differentiation, c-JUN is initially downregulated but later reactivated

    • These temporal dynamics may explain seemingly contradictory functions at different stages

This functional versatility of c-JUN likely explains its evolutionary conservation—it serves as a flexible transcriptional regulator that can be repurposed for different functions across cell types and developmental contexts. The core DNA binding and dimerization domains are conserved, while the regulatory mechanisms and interaction partners have evolved to enable diverse functions.

In research settings, these contradictions highlight the importance of studying c-JUN in appropriate cellular contexts rather than extrapolating findings across cell types.

What are the most promising future research directions in c-JUN biology?

Several promising research directions in c-JUN biology are emerging that could significantly advance our understanding of this transcription factor and its therapeutic potential:

  • Single-cell approaches to c-JUN function:

    • Single-cell transcriptomics and epigenomics to understand cell-to-cell variation in c-JUN activity

    • Spatial transcriptomics to map c-JUN activity in complex tissues

    • These approaches could resolve contradictory findings by identifying cell subpopulations with distinct c-JUN functions

  • Structural biology of c-JUN complexes:

    • Cryo-EM structures of c-JUN in complex with different dimerization partners

    • Structural characterization of the c-JUN IRES element in glioblastoma

    • Structure-based drug design targeting specific c-JUN interactions

  • Therapeutic targeting refinement:

    • Development of selective inhibitors of specific c-JUN functions

    • Targeted degradation approaches (e.g., PROTACs) specific to c-JUN

    • Combination therapies that address resistance mechanisms

  • Translational regulation mechanisms:

    • Further characterization of IRES-mediated translation of c-JUN in different cancers

    • Investigation of RNA binding proteins regulating c-JUN translation

    • Potential role of RNA modifications in regulating c-JUN mRNA translation

  • Systems biology approaches:

    • Network modeling of c-JUN interactions across cell types

    • Integration of multi-omics data to build predictive models of c-JUN function

    • Computational approaches to predict context-specific c-JUN targets

  • c-JUN in cell fate engineering:

    • Leveraging c-JUN manipulation to improve directed differentiation protocols

    • Engineering c-JUN activity to enhance CAR T cell function in solid tumors

    • Reprogramming approaches based on modulating c-JUN activity

  • Evolutionary studies of AP-1 regulation:

    • Comparative analysis of c-JUN regulation across species

    • Understanding how context-dependent functions evolved

    • Identifying conserved versus divergent regulatory mechanisms

Product Science Overview

Introduction

The Jun proto-oncogene, also known as c-Jun, is a crucial component of the AP-1 transcription factor complex. This gene plays a significant role in regulating gene expression in response to various stimuli, including stress, cytokines, growth factors, and infections. The human recombinant form of this protein is often used in research to study its functions and interactions.

Gene and Protein Structure

The Jun proto-oncogene is located on chromosome 1p32-p31 in humans . It encodes a protein that is highly similar to the viral oncoprotein v-jun, discovered in avian sarcoma virus 17 . The human JUN gene is intronless, meaning it does not contain introns within its coding sequence .

The protein encoded by the JUN gene is a part of the AP-1 transcription factor complex, which also includes proteins like c-Fos. This complex binds to specific DNA sequences to regulate the transcription of various genes .

Function and Regulation

The primary function of the Jun protein is to regulate gene expression by binding to the AP-1 binding site on DNA. This regulation is crucial for various cellular processes, including cell proliferation, differentiation, and apoptosis .

Jun and its dimerization partners in the AP-1 complex are regulated by diverse extracellular stimuli, such as peptide growth factors, pro-inflammatory cytokines, oxidative stress, and UV irradiation . For instance, UV irradiation is a potent inducer of elevated c-Jun expression . The transcription of c-Jun can be autoregulated by its own product, creating a positive feedback loop that prolongs the signals from extracellular stimuli .

Role in Cancer

The Jun proto-oncogene is classified as a proto-oncogene because its deregulation can lead to cancer. It was the first oncogenic transcription factor discovered . The protein’s activity is regulated by the ERK pathway, and constitutively active ERK can increase c-Jun transcription and stability . This regulation is significant for the activity of c-Jun in cancer, as it can lead to the activation of downstream targets like RACK1 and cyclin D1, which are involved in cell cycle progression and tumorigenesis .

Research and Applications

Human recombinant Jun protein is widely used in research to study its role in gene regulation and its implications in cancer. By using the recombinant form, researchers can investigate the protein’s interactions, regulatory mechanisms, and potential as a therapeutic target.

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 2025 TheBiotek. All Rights Reserved.