C-JUN Human (241 a.a.)

Jun Proto-Oncogene (1-241 a.a.) Human Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Composition

  • Sequence: 261 amino acids (1–241 native residues + 20-amino acid His tag) .

  • Gene Location: Chromosome 1p32-p31 (intronless) .

  • Domains:

    • Transactivation domain (aa 6–194) .

    • Basic DNA-binding domain (aa 257–276 in human c-Jun) .

    • Leucine zipper dimerization motif .

Key Interactions

  • Forms heterodimers with FOS family proteins (e.g., c-Fos) to create the AP-1 transcription factor complex .

  • Binds AP-1 consensus sequences (5'-TGA[GC]TCA-3') to regulate target genes .

  • Interacts with PU.1 and C/EBPβ in macrophages to coactivate inflammatory genes like IL-1β without direct DNA binding .

Transcriptional Regulation

  • AP-1 Activation: Drives expression of genes involved in cell cycle progression (e.g., cyclin D1), apoptosis (e.g., FASLG), and inflammation (e.g., COX2) .

  • Dual Phosphorylation: Phosphorylation at Ser63/73 and Thr91/93 by JNK enhances transcriptional activity .

Disease Associations

DiseaseRole of c-JunKey Findings
CancerPro-oncogenic in breast and lung cancers; tumor-suppressive in KRAS-driven tumorsOverexpression accelerates metastasis , while deletion increases lung tumor burden .
ArthritisPromotes inflammation via COX2 upregulation and ARG1 suppressionc-Jun-deficient mice show reduced joint inflammation and bone erosion .
NeurodegenerationMediates TE mobilization in Alzheimer’s diseaseUpregulation in AD models triggers genomic instability via cGAS-STING activation .
DiabetesInduces β-cell apoptosis via JNK/AP-1 signalingHuman amylin activates c-Jun, leading to caspase-dependent apoptosis .

Experimental Models

  • In Vitro Studies:

    • Used in electrophoretic mobility shift assays (EMSAs) to assess AP-1 DNA-binding activity .

    • Overexpressed in MCF-7 breast cancer cells to study tumor aggression .

  • In Vivo Models:

    • Conditional knockout mice (c-Jun ΔLysM/ΔMx) reveal roles in arthritis and T-cell development .

Therapeutic Targeting

  • Inhibitors: Tylophorine (plant alkaloid) induces G1 arrest by downregulating c-Jun targets like cyclin A2 .

  • Gene Therapy: Antisense c-jun (AS-c-jun) reduces apoptosis in β-cells .

Controversies and Emerging Insights

  • Context-Dependent Roles:

    • Oncogene vs. Tumor Suppressor: c-Jun promotes metastasis in breast cancer but suppresses KRAS-driven lung tumors .

    • JunD Compensation: In c-Jun-deficient cells, JunD overexpression rescues AP-1 activity, highlighting functional redundancy .

  • Phosphorylation Dynamics:

    • Heterodimerization with ATF2 prevents nuclear export, enhancing transcriptional output .

Key Research Findings

  1. Macrophage Polarization: c-Jun deletion shifts macrophages toward anti-inflammatory states (↑Arg1, ↓Cox2), mitigating arthritis .

  2. Neurodegeneration: c-Jun upregulation in Alzheimer’s models correlates with transposable element mobilization and neuroinflammation .

  3. T-Cell Development: c-Jun regulates αβ/γδ T-cell lineage choice by suppressing IL-7Rα expression .

Future Directions

  • Therapeutic Strategies: Targeting c-Jun-JNK interactions in inflammatory diseases .

  • Systems Biology: Elucidating c-Jun’s role in chromatin remodeling and 3D genomic architecture .

Product Specs

Introduction
C-JUN, in conjunction with c-Fos, forms the AP-1 early response transcription factor. Activated by the JNK pathway, C-JUN is recognized as the likely transforming gene of avian sarcoma virus 17. This protein exhibits high similarity to the viral protein and directly interacts with specific DNA target sequences to regulate gene expression. Notably, the C-JUN gene lacks introns and maps to chromosome region 1p32-p31, an area often associated with translocations and deletions in human cancers.
Description
Recombinant human C-JUN, expressed in E. coli, is fused with a 20 amino acid His tag at its N-terminus. This results in a single, non-glycosylated polypeptide chain comprising 261 amino acids (1-241 a.a.) with a molecular weight of 27.3 kDa. Purification of C-JUN is achieved using proprietary chromatographic techniques.
Physical Appearance
Clear, colorless, and sterile-filtered solution.
Formulation
The C-JUN solution is provided at a concentration of 0.25 mg/ml in a buffer consisting of 20 mM Tris-HCl (pH 8.0), 10% glycerol, 2 mM DTT, and 0.1 M NaCl.
Stability
For short-term storage (up to 2-4 weeks), the product can be stored at 4°C. For extended storage, freezing at -20°C is recommended. To further enhance stability during long-term storage, adding a carrier protein like HSA or BSA (0.1%) is advised. Repeated freeze-thaw cycles should be avoided.
Purity
Purity exceeds 90% as determined by SDS-PAGE analysis.
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.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MTAKMETTFY DDALNASFLP SESGPYGYSN PKILKQSMTL NLADPVGSLK PHLRAKNSDL LTSPDVGLLK LASPELERLI IQSSNGHITT TPTPTQFLCP KNVTDEQEGF AEGFVRALAE LHSQNTLPSV TSAAQPVNGA GMVAPAVASV AGGSGSGGFS ASLHSEPPVY ANLSNFNPGA LSSGGGAPSY GAAGLAFPAQ PQQQQQPPHH LPQQMPVQHP RLQALKEEPQ TVPEMPGETP P.

Q&A

What is the functional significance of C-JUN in human cell development?

C-JUN acts on chromatin loci to influence cell fate specification, particularly as cells exit pluripotency. Although widely expressed across various cell types in early embryogenesis, C-JUN is not essential for maintaining pluripotency. Instead, it functions as a repressor to constrain mesoderm development while having minimal impact on ectoderm differentiation .

This context-dependent activity is critical for proper embryonic development, as C-JUN interacts with specific protein complexes to regulate chromatin accessibility at developmental genes. The protein's role varies significantly between differentiation pathways, making it an important factor in understanding lineage commitment mechanisms.

How does C-JUN function in transcriptional regulation?

C-JUN can operate through two distinct mechanisms:

  • As a DNA-binding activator: C-JUN binds directly to AP-1 sequences to drive gene expression

  • As a coactivator: C-JUN functions as a cofactor for other transcription factors (like PU.1 in macrophages) without direct DNA binding

The mechanism whereby C-JUN switches between these modes involves its basic domain, which can either interact with DNA or participate in protein-protein interactions. Notably, C-JUN homodimers bind AP-1 sequences with lower affinity than heterodimers formed with other AP-1 family members . When functioning as a coactivator, C-JUN enhances transcription by facilitating the assembly of the RNA polymerase II preinitiation complex with minimal effect on local chromatin status .

What structural features enable C-JUN's dual functionality?

Human C-JUN consists of 241 amino acids in its native form and contains several key functional domains:

  • Basic DNA-binding domain: Contains four critical residues (positions R270, N271, C278, R279) that can either contact DNA or participate in protein-protein interactions

  • Leucine zipper dimerization domain: Enables formation of homo- and heterodimers

  • Transactivation domain: Contains phosphorylation sites regulated by the JNK pathway

The recombinant form often used in research includes a 20-amino acid His tag at the N-terminus, resulting in a 261-amino acid protein with a molecular mass of 27.3kDa . The fact that the same basic domain residues mediate both DNA binding and protein interactions creates a mutually exclusive switch between C-JUN's functions as a direct transcription factor and as a coactivator .

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

C-JUN acts as a chromatin repressor that specifically limits mesoderm differentiation during human pluripotent stem cell differentiation through several mechanisms:

  • C-JUN interacts with the MBD3-NuRD complex to maintain chromatin in a low accessibility state at mesoderm-related genes

  • This interaction specifically inhibits the activation of key mesoderm factors such as EOMES and GATA4

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

  • Conversely, overexpressing C-JUN redirects differentiation toward a fibroblast-like lineage

Experimental evidence shows that in a 3-day mesoderm induction system using CHIR99021 (WNT pathway activator), C-JUN knockout cells show increased proportions of PDGFRA+ mesoderm cells compared to wild-type cells. This phenotype can be replicated using the JNK inhibitor SP600125 .

What is the relationship between C-JUN and the MBD3-NuRD complex?

C-JUN and the MBD3-NuRD complex have a complex, context-dependent relationship:

  • C-JUN can directly interact with MBD3, a component of the NuRD complex

  • This interaction recruits the NuRD complex (a co-repressor that mediates gene silencing through histone deacetylation and chromatin remodeling) to target genes

  • The interaction is disrupted by C-JUN N-terminal phosphorylation, which is induced by JNK signaling

  • Knockdown of MBD3 enhances mesoderm generation, whereas MBD3 overexpression impedes it

The dynamic nature of this interaction provides a mechanism for fine-tuning gene expression during development. When C-JUN is phosphorylated, its interaction with MBD3 is disrupted, potentially alleviating repression of mesoderm genes .

How does C-JUN antagonize WNT signaling during mesoderm formation?

WNT signaling plays a pivotal role in mesoderm specification and cardiac development. C-JUN functions as an antagonist to WNT signaling during the transition from pluripotent stem cells to mesoderm through the following mechanisms:

  • C-JUN is rapidly degraded following CHIR99021 treatment (WNT activator), with a concomitant decrease in mRNA levels

  • Deletion of C-JUN leads to upregulation of β-CATENIN expression, enhancing WNT signaling and expediting mesoderm formation

  • Elevated C-JUN expression suppresses β-CATENIN and redirects cell fate toward a fibroblast-like state

This antagonistic relationship explains previous observations that C-JUN knockout enhances cardiac generation (mesoderm-derived), while C-JUN overexpression inhibits this process .

What techniques are most effective for studying C-JUN's dual functionality?

To investigate C-JUN's role as both a DNA-binding transcription factor and a coactivator, researchers should consider:

  • Mutational analysis: Create point mutations in C-JUN's basic domain:

    • R270I/N271D (M13) mutation

    • C278D/R279I (M14b) mutation

    • Double mutant combining both mutations (M13-14b)

  • Protein-protein interaction assays:

    • Co-immunoprecipitation with HA-tagged C-JUN constructs

    • GST pull-down assays using GST-C/EBPβ and GST-PU.1 fusion constructs

    • In vitro binding assays with purified proteins

  • DNA binding assays:

    • Electrophoretic mobility shift assays (EMSA) to assess direct DNA binding

    • Chromatin immunoprecipitation (ChIP) to identify genomic binding sites

  • Functional validation:

    • Reporter gene assays using AP-1 responsive elements

    • Analysis of target gene expression in the presence of wildtype vs. mutant C-JUN

These approaches allow for distinguishing between C-JUN's direct transcriptional effects and its coactivator functions in different cellular contexts.

What are optimal methods for C-JUN knockout and inhibition studies?

Based on the research methodologies described in the search results, effective approaches include:

  • Genetic manipulation:

    • CRISPR/Cas9-mediated knockout of C-JUN in human pluripotent stem cells

    • Generation of multiple independent knockout clones (e.g., C-JUN−/− H1 #2 and #10)

    • Validation of normal karyotype and pluripotency markers in knockout lines

  • Pharmacological inhibition:

    • JNK inhibitor (SP600125) treatment to prevent C-JUN phosphorylation and activation

    • Dose-dependent inhibition studies to establish optimal concentrations

    • Time-course experiments to determine temporal requirements for inhibition

  • Validation approaches:

    • Western blotting to confirm protein absence/inhibition

    • RT-qPCR to assess effects on target gene expression

    • Flow cytometry for lineage markers (e.g., PDGFRA+ for mesoderm)

    • Transcriptome analysis to assess global effects on gene expression

Combining genetic and pharmacological approaches provides complementary evidence and controls for potential off-target effects of either method.

How can researchers effectively produce and purify recombinant C-JUN protein?

To obtain high-quality recombinant human C-JUN protein for in vitro studies:

  • Expression system:

    • E. coli expression system for non-glycosylated protein

    • Human C-JUN (amino acids 1-241) with N-terminal 20-amino acid His tag

    • Expected molecular mass of 27.3kDa

  • Purification approach:

    • Multi-step chromatographic techniques

    • Immobilized metal affinity chromatography (IMAC) utilizing the His tag

    • Additional purification steps may include ion exchange and size exclusion chromatography

    • Final product should be a sterile filtered colorless solution

  • Quality control:

    • SDS-PAGE to confirm purity and correct molecular weight

    • Western blotting with anti-C-JUN antibodies

    • Functional assays to confirm DNA binding activity

    • Mass spectrometry to verify protein identity

For functional studies, researchers should consider whether post-translational modifications absent in E. coli-expressed protein might affect activity in specific experimental contexts.

How should researchers interpret contradictory findings about C-JUN function across different model systems?

When faced with seemingly contradictory results regarding C-JUN function:

  • Consider species-specific differences:

    • C-JUN knockout in mice results in embryonic lethality around E13.5

    • Human and mouse C-JUN may have different roles due to distinct evolutionary paths

    • The same genes may play varied roles across species due to differences in developmental processes

  • Evaluate cellular context:

    • C-JUN has opposing effects in different cell types (inhibits mesoderm but minimal effect on ectoderm)

    • Temporal dynamics of C-JUN expression affect its function (initial downregulation followed by reactivation during mesoderm differentiation)

    • Interactions with different protein partners may alter C-JUN function

  • Examine methodology differences:

    • Complete knockout versus partial knockdown

    • Constitutive versus inducible/temporal manipulation

    • Different differentiation protocols or culture conditions

  • Analyze signaling pathway crosstalk:

    • C-JUN antagonizes WNT signaling during mesoderm differentiation

    • JNK signaling affects C-JUN phosphorylation state and interaction with partners like MBD3

A comprehensive evaluation considering these factors helps reconcile apparent contradictions and build a more nuanced understanding of C-JUN biology.

What bioinformatic approaches are most valuable for analyzing C-JUN genomic targets?

For comprehensive analysis of C-JUN genomic targets:

  • Integrated genomic approaches:

    TechniqueApplicationData Output
    ChIP-seqIdentify C-JUN binding sitesGenome-wide binding profile
    ATAC-seqAssess chromatin accessibilityOpen chromatin regions
    RNA-seqMeasure gene expression changesDifferential expression
    CUT&RUNHigher resolution bindingPrecise binding locations
    HiC/3CChromatin interactionsLong-range regulatory contacts
  • Motif analysis:

    • Identify enriched AP-1 motifs in C-JUN binding sites

    • Discover co-occurring motifs for potential interaction partners

    • Compare motif enrichment between different cell types/conditions

  • Integration with protein interaction data:

    • Correlate genomic binding with known protein-protein interactions

    • Identify regions where C-JUN may function as a coactivator versus direct DNA binder

    • Map binding sites to the MBD3-NuRD complex to identify repressed loci

These approaches provide a comprehensive view of how C-JUN regulates gene expression across the genome in different cellular contexts.

How might modulation of C-JUN activity be leveraged for directed differentiation in regenerative medicine?

Based on C-JUN's role in mesoderm differentiation, strategic manipulation could enhance regenerative medicine applications:

  • Cardiac regeneration:

    • Temporary inhibition of C-JUN (using JNK inhibitors or genetic approaches) could enhance mesoderm differentiation and subsequent cardiomyocyte generation

    • Temporal modulation might be required to match developmental sequences

    • This approach could improve efficiency of cardiac differentiation protocols for cell therapy or tissue engineering

  • Prevention of fibrosis:

    • Since C-JUN overexpression redirects differentiation toward fibroblast-like lineages, inhibiting C-JUN during tissue regeneration might reduce fibrotic scarring

    • This could be particularly relevant for cardiac repair after myocardial infarction

  • Balancing differentiation pathways:

    • C-JUN inhibition could be combined with pathway activators (e.g., WNT modulators) for synergistic effects on mesoderm differentiation

    • Sequential modulation of C-JUN at different stages might optimize differentiation outcomes

  • Small molecule approaches:

    • JNK inhibitors like SP600125 offer a pharmacological approach to inhibit C-JUN activity

    • Development of compounds targeting the C-JUN/MBD3 interaction could provide more specific modulation

Understanding the precise temporal requirements for C-JUN inhibition during differentiation is essential for translating these findings to clinical applications.

What are the potential implications of C-JUN research for understanding developmental disorders?

C-JUN's role in early embryonic development suggests several implications for developmental disorders:

  • Congenital heart defects:

    • Since C-JUN regulates mesoderm differentiation and cardiac development, dysregulation might contribute to congenital heart defects

    • Studying C-JUN interactions in patient-derived iPSCs could reveal mechanisms underlying these conditions

  • Developmental signaling disorders:

    • C-JUN's antagonism of WNT signaling connects it to disorders involving WNT pathway dysregulation

    • Aberrant interactions between C-JUN and the MBD3-NuRD complex could affect chromatin regulation during development

  • Human-specific developmental processes:

    • Research on human C-JUN is particularly valuable given the differences between mouse and human development

    • Findings may explain human-specific aspects of developmental disorders not captured in animal models

  • Disease modeling approaches:

    • Patient-derived iPSCs with genetic variants affecting C-JUN or its pathway components

    • CRISPR-engineered mutations in C-JUN interaction domains

    • Small molecule modulators of C-JUN activity to assess developmental phenotypes

These research directions could provide insights into pathogenic mechanisms and potential therapeutic approaches for developmental disorders.

Product Science Overview

Introduction

The Jun Proto-Oncogene, also known as c-Jun, is a crucial component of the Activator Protein 1 (AP-1) transcription factor complex. This protein plays a significant role in regulating gene expression in response to a variety of stimuli, including stress, cytokines, growth factors, and infections.

Structure and Production

The human recombinant Jun Proto-Oncogene (1-241 a.a.) is a non-glycosylated polypeptide chain consisting of 261 amino acids, including a 20 amino acid His tag at the N-terminus . This recombinant protein is produced in Escherichia coli (E. coli) and has a molecular mass of approximately 27.3 kDa .

Function and Mechanism

c-Jun is highly similar to the viral protein and interacts directly with specific target DNA sequences to regulate gene expression . It is activated by the c-Jun N-terminal kinase (JNK) pathway and forms a part of the AP-1 early response transcription factor in combination with c-Fos . The c-Jun gene is intronless and is mapped to the chromosomal region 1p32-p31, which is involved in both translocations and deletions in human malignancies .

Applications

The recombinant form of c-Jun is widely used in laboratory research to study gene expression regulation, signal transduction pathways, and the molecular mechanisms underlying various diseases, including cancer . It is also utilized in drug development and screening processes.

Storage and Stability

The c-Jun recombinant protein is typically stored at 4°C if it will be used within 2-4 weeks. For longer storage periods, it is recommended to keep it frozen at -20°C with the addition of a carrier protein such as 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 2025 TheBiotek. All Rights Reserved.