FOSL2 Human

FOS-Like Antigen 2 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to FOSL2 Human

FOSL2 (FOS-like antigen 2), encoded by the FOSL2 gene on human chromosome 2 (positions 28,392,448–28,417,317), is a transcription factor critical for regulating cellular responses to stimuli, including proliferation, differentiation, and inflammation . As a member of the FOS gene family (c-Fos, FOSB, FOSL1, FOSL2), it dimerizes with JUN proteins to form the AP-1 transcription factor complex, which binds DNA to modulate gene expression . Below, we detail its structural, functional, and clinical implications, supported by recent research findings.

Gene and Protein Structure of FOSL2

FeatureDetailsSource
Chromosomal LocationChromosome 2 (28,392,448–28,417,317 forward strand)
Transcripts4 splice variants (ENST00000264716, ENST00000379619, etc.)
Protein Length326 amino acids (canonical isoform)
Molecular Mass37.6 kDa (recombinant protein expressed in E. coli)
Key DomainsLeucine zipper for dimerization with JUN proteins

Notes:

  • The canonical isoform (ENST00000264716.9) encodes a 326-amino acid protein with a His-tag in recombinant forms .

  • Splice variants include truncated forms (e.g., 202 aa and 202 aa variants) .

Regulation of Leptin Expression

FOSL2 directly activates leptin (LEP) transcription in adipocytes by binding a cis-element upstream of the LEP gene. Key findings:

  • Human Adipocytes: Knockdown reduces LEP mRNA by ~50% .

  • Mouse Models: Overexpression in adipocytes increases Lep levels, while adipocyte-specific deletion reduces Lep expression .

  • Obesity Correlation: Elevated FOSL2 levels in obese mice and humans correlate with higher LEP expression .

Immunosuppression in Pancreatic Ductal Adenocarcinoma (PDAC)

FOSL2 promotes PDAC progression via the KRAS/MAPK pathway:

  • Mechanism:

    • KRAS activation upregulates FOSL2 .

    • FOSL2 transcriptionally activates CCL28, recruiting regulatory T (Treg) cells to create an immunosuppressive microenvironment .

  • Clinical Relevance: High FOSL2 expression correlates with poor prognosis in PDAC patients .

Modulation of Th17 Cell Differentiation

FOSL2 interacts with RNA-binding proteins (RBPs) to regulate Th17 cell function:

  • Interactome: Shared binding partners with FOSL1 include JUN proteins, XRN1/XRN2 (mRNA decay), and RORγT (Th17 master regulator) .

  • Functional Impact: FOSL2 negatively regulates Th17 differentiation by destabilizing lineage-specific transcripts via RBP complexes .

Aging and Tissue Regeneration

FOSL2 expression declines with age in bone marrow-derived mesenchymal stromal cells (hMSCs):

  • Correlation: Linear regression shows a negative slope (-0.02442) between age and FOSL2 expression (P = 0.00081) .

  • Implications: Reduced FOSL2 may contribute to diminished regenerative capacity in aging hMSCs .

Comparative Analysis of FOSL2 Roles

Biological ProcessMechanismKey FindingsSource
Leptin RegulationBinds LEP enhancer, activates transcriptionObesity-linked FOSL2 upregulation drives LEP
PDAC ProgressionKRAS → FOSL2 → CCL28 → Treg recruitmentFOSL2 depletion reduces tumor growth
Th17 Cell FateAssociates with RBPs to destabilize mRNAFOSL2 knockdown enhances Th17 differentiation
AgingDeclining expression in hMSCsCorrelates with reduced regenerative capacity

Research and Therapeutic Implications

FOSL2’s multifaceted roles highlight its potential as a therapeutic target:

  • Obesity/Diabetes: Inhibiting FOSL2 could suppress leptin overproduction in obesity .

  • Cancer Immunotherapy: Targeting FOSL2-CCL28 axis may enhance anti-tumor immunity in PDAC .

  • Aging and Regeneration: Modulating FOSL2 expression could improve hMSC functionality in regenerative medicine .

Product Specs

Introduction
FOS-Like Antigen 2, also known as FOSL2, is a protein encoded by the FOSL2 gene in humans. The Fos gene family comprises four members: FOS, FOSB, FOSL1, and FOSL2. These genes encode leucine zipper proteins capable of dimerizing with proteins from the JUN family, forming the AP-1 transcription factor complex. Consequently, FOS proteins are implicated in regulating cell proliferation, differentiation, and transformation.
Description
Recombinant human FOSL2, produced in E. coli, is a single, non-glycosylated polypeptide chain consisting of 349 amino acids (1-326 a.a.). It has a molecular weight of 37.6 kDa. A 23-amino acid His-tag is fused to the N-terminus of FOSL2, and the protein is purified using proprietary chromatographic techniques.
Physical Appearance
A clear, sterile-filtered solution.
Formulation
The FOSL2 protein solution has a concentration of 0.5 mg/ml and is supplied in a buffer containing 20mM Tris-HCl (pH 8.0) and 10% glycerol.
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For extended storage, freeze the product at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Repeated freezing and thawing should be avoided.
Purity
The purity of FOSL2 is determined to be greater than 85.0% by SDS-PAGE analysis.
Synonyms
FOS-Like Antigen 2, FRA-2, FRA2, Fos-Related Antigen 2, FOSL2.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMYQDYPG NFDTSSRGSS GSPAHAESYS SGGGGQQKFR VDMPGSGSAF IPTINAITTS QDLQWMVQPT VITSMSNPYP RSHPYSPLPG LASVPGHMAL PRPGVIKTIG TTVGRRRRDE QLSPEEEEKR RIRRERNKLA AAKCRNRRRE LTEKLQAETE ELEEEKSGLQ KEIAELQKEK EKLEFMLVAH GPVCKISPEE RRSPPAPGLQ PMRSGGGSVG AVVVKQEPLE EDSPSSSSAG LDKAQRSVIK PISIAGGFYG EEPLHTPIVV TSTPAVTPGT SNLVFTYPSV LEQESPASPS ESCSKAHRRS SSSGDQSSDS LNSPTLLAL

Q&A

What is FOSL2 and what is its fundamental role in human cellular processes?

FOSL2 functions as a transcription factor within the activator protein-1 (AP-1) complex, orchestrating cellular responses to various stimuli including immune surveillance and tissue-resident memory T cell differentiation. It primarily operates by forming heterodimers with JUN proteins to regulate gene expression . Research demonstrates that FOSL2 has tissue-specific functions across different cell types, with significant roles in Th17 cell differentiation where it modulates effector functions through complex transcriptional networks .

FOSL2 binds to specific DNA sequences in promoter and enhancer regions, influencing the transcription of target genes involved in cell proliferation, differentiation, and immune regulation. Experimental data from immunoblot analysis reveals upregulation of FOSL2 during Th17 cell differentiation, with levels significantly increased compared to activated (Th0) cells after 72 hours of differentiation .

How is FOSL2 expression regulated throughout the human lifespan?

FOSL2 expression demonstrates significant age-dependent regulation in human tissues. Quantitative research using qPCR and RNA sequencing in human bone marrow-derived mesenchymal stromal cells (hMSCs) from donors aged 17-84 years reveals a statistically significant negative correlation between FOSL2 expression and age .

Statistical analysis shows:

  • Slope: -0.02442

  • R-value: -0.41759

  • P-value: 0.00081

This significant inverse relationship indicates that FOSL2 expression reliably decreases with advancing age . The transcriptional changes in FOSL2 and other AP-1 complex members emerge as a conserved signature of immune aging, potentially contributing to increased inflammation and senescence, a phenomenon often termed "inflammaging" .

What experimental approaches are most effective for measuring FOSL2 expression in human samples?

Measuring FOSL2 expression in clinical samples requires sophisticated methodological approaches:

RNA-based methods:

  • Quantitative PCR (qPCR): Successfully employed to measure FOSL2 mRNA expression across different ages in human bone marrow-derived mesenchymal stromal cells .

  • RNA Sequencing: Provides comprehensive transcriptomic analysis that can detect splice variants and contextual expression patterns .

Protein-based methods:

  • Immunoblot analysis: Used effectively to detect FOSL2 protein levels during Th17 cell differentiation .

  • Immunohistochemistry (IHC): Valuable for spatial visualization in heterogeneous tissues like tumors .

Epigenetic approaches:

  • ATAC-seq: Assesses chromatin accessibility at FOSL2 regulatory regions .

  • H3K27ac ChIP-seq: Identifies active enhancers associated with FOSL2 .

For microarray experiments, gene expression is typically quantified as relative expression levels derived from fluorescence intensity, with raw values transformed into logarithmic scale (usually log2) to stabilize variance and enhance interpretability .

What is the role of FOSL2 in the pathogenesis of pancreatic ductal adenocarcinoma?

FOSL2 plays a crucial role in pancreatic ductal adenocarcinoma (PDAC) progression through multiple mechanisms. Research using genetically engineered mouse models (GEMMs) with comprehensive multiomics sequencing (ATAC-seq, H3K27ac ChIP-seq, and RNA-seq) has identified FOSL2 as significantly upregulated in PDAC and associated with poor patient prognosis .

Functional studies demonstrate that FOSL2 promotes:

  • Enhanced cancer cell proliferation

  • Increased migration capability

  • Augmented invasion potential

Mechanistically, FOSL2 operates downstream of the KRAS/MAPK pathway, which is frequently activated in PDAC due to KRAS mutations. The research reveals that FOSL2 transcriptionally activates C-C motif chemokine ligand 28 (CCL28), which recruits immunosuppressive regulatory T (Treg) cells to the tumor microenvironment . This establishes a distinct immunosuppressive regulatory axis:

KRAS mutation → MAPK activation → FOSL2 upregulation → CCL28 transcription → Treg recruitment → Immunosuppression → Enhanced tumor progression

These findings highlight FOSL2 as a critical mediator between oncogenic KRAS signaling and immune evasion in pancreatic cancer.

How does the FOSL2 interactome function in human Th17 cells?

The FOSL2 interactome in human Th17 cells reveals complex protein interaction networks that regulate immune function. Research using affinity purification-mass spectrometry analysis has established the first comprehensive map of FOSL2 protein interactions in these cells .

Key findings from interactome studies include:

  • Beyond known JUN protein partnerships, numerous novel binding partners of FOSL2 have been identified .

  • Gene ontology analysis revealed that a significant fraction of these interactors are associated with RNA-binding activity, suggesting unexpected mechanistic links between transcriptional and post-transcriptional regulation .

  • Twenty-nine proteins were found to share interactions with both FOSL1 and FOSL2, including key regulators of Th17 cell fate .

  • The interactome includes RNA-binding proteins and exonucleases like XRN1 and XRN2, along with their partners UPF1 and UPF2, which trigger mRNA decay .

The detection of RNA processing factors in the FOSL2 interactome suggests that beyond direct transcriptional regulation, FOSL2 might influence gene expression through post-transcriptional mechanisms, potentially restraining Th17 signaling by associating with proteins that destabilize lineage-specific transcripts .

What are the molecular mechanisms linking KRAS mutations and FOSL2 activity in cancer?

The relationship between KRAS mutations and FOSL2 represents a critical signaling axis in cancer pathogenesis. KRAS mutations, which occur in approximately 90% of pancreatic ductal adenocarcinoma cases, lead to constitutive activation of the MAPK signaling pathway. Research employing multiomics approaches has demonstrated that this activation results in increased expression and activity of FOSL2, positioning it as a key downstream effector of oncogenic KRAS signaling .

The molecular cascade follows this sequence:

  • KRAS mutation → Constitutive MAPK pathway activation

  • MAPK pathway activation → Increased FOSL2 expression

  • Elevated FOSL2 → Transcriptional activation of target genes including CCL28

  • CCL28 secretion → Recruitment of immunosuppressive Treg cells

  • Immunosuppressive microenvironment → Enhanced tumor progression

Studies employing Cleavage Under Targets and Tagmentation (CUT&Tag) have identified direct genomic targets of FOSL2, clarifying its role as a transcriptional regulator downstream of KRAS . Interestingly, research in glioblastoma suggests that FOSL2 may also play broader roles in cancer beyond specific mutational contexts, as the enrichment of the FOSL2 regulon does not correlate with a specific GBM mutational subtype .

What techniques are most effective for studying FOSL2 protein-protein interactions?

Studying FOSL2 protein interactions requires sophisticated methodological approaches:

Affinity Purification coupled with Mass Spectrometry (AP-MS):
This approach has been successfully employed to establish FOSL2 interactomes in human Th17 cells through:

  • Immunoprecipitation using specific antibodies against FOSL2

  • Mass spectrometric analysis of co-precipitated proteins

  • Computational filtering using the MiST algorithm to calculate scores based on intensity, consistency, and specificity

  • Data filtering to retain proteins with three valid values in at least one group (IgG, FOSL1, and FOSL2 pull-down)

Validation techniques:

  • Parallel Reaction Monitoring (PRM) targeted mass spectrometry

  • Comparison with protein databases to eliminate common contaminants (proteins detected with frequency <40% in other IP experiments)

Network analysis tools:

  • STRING database mapping and Cytoscape visualization for protein-protein interaction networks

  • Perseus software for statistical analysis and visualization of data

The research data has been made publicly available through repositories including PRIDE (identifier PXD025729) and the Skyline Panorama (https://panoramaweb.org/FOSL1_2_Th17.url)[1], enabling validation and extension of findings by other researchers.

What are the best experimental models for studying FOSL2 function in human disease contexts?

Selecting appropriate experimental models is crucial for investigating FOSL2 function in disease:

Cell-based systems:

  • Primary human CD4+ T cells: Utilized for studying FOSL2 in Th17 differentiation, with cells activated and differentiated toward Th17 fate for 72 hours .

  • Human bone marrow-derived mesenchymal stromal cells (hMSCs): Employed for studying age-dependent FOSL2 expression changes using samples from donors aged 17-84 years .

  • Cancer cell lines: Valuable for mechanistic studies with defined genetic backgrounds.

Animal models:

  • Genetically Engineered Mouse Models (GEMMs): Models with or without KRAS and/or TP53 mutations have been effectively used to study FOSL2's role in pancreatic cancer .

Functional assays:

  • Proliferation assays (e.g., CCK8)

  • Migration and invasion assays (e.g., transwell)

  • ChIP-qPCR and dual-luciferase reporter assays to determine direct transcriptional targets

Multi-omics approaches:

  • Epigenomic profiling: ATAC-seq and H3K27ac ChIP-seq to characterize the epigenetic landscape

  • Transcriptomic analysis: RNA-seq to identify expression changes

  • Proteomic methods: Mass spectrometry for protein quantification

  • Interactome mapping: AP-MS, CUT&Tag

For glioma research, consensus clustering of larger aggregated datasets like GBMap has generated more biologically plausible results compared to smaller datasets .

How can researchers effectively use FOSL2 gene expression data to understand age-related changes in human tissues?

When analyzing FOSL2 expression in relation to aging, several methodological considerations are essential:

Study design considerations:

  • Cross-sectional studies examining FOSL2 expression across different age groups should acknowledge limitations in capturing individual longitudinal changes .

  • Sample size calculations should account for the expected effect size based on previous studies (approximately -0.02442 expression units per year of age) .

Statistical approaches:

  • Linear regression analysis has been effectively employed to assess the relationship between FOSL2 expression and donor age, generating correlation coefficients and p-values to evaluate statistical significance .

  • Data visualization using scatter plots with regression lines helps illustrate the negative correlation between FOSL2 expression and age .

Data normalization:

  • Raw intensity values should be transformed into logarithmic scale (typically log2) to stabilize variance and enhance interpretability .

  • Proper normalization procedures must be applied to correct for background noise and inter-array variations when using microarray data .

Contextual analysis:

  • FOSL2 expression should be analyzed in relation to other aging-associated genes to identify coordinated expression networks .

  • The connection between FOSL2 expression changes and functional alterations in tissue regenerative capacity should be explored through pathway analysis .

When utilizing publicly available datasets like GSE39540 (containing data from 61 donors aged 17-84 years), researchers should explicitly document the platform used (e.g., Affymetrix Human Genome U133A 2.0 Array) and provide comprehensive methodology for reproducibility .

How can researchers distinguish between correlation and causation when studying FOSL2 in disease pathogenesis?

Establishing causality in FOSL2 research requires multiple complementary approaches:

Genetic manipulation strategies:

  • Loss-of-function studies: Knockdown or knockout of FOSL2 using siRNA, shRNA, or CRISPR-Cas9 technologies to demonstrate necessity.

  • Gain-of-function studies: Overexpression of wild-type or mutant FOSL2 to establish sufficiency.

  • Rescue experiments: Reintroduction of FOSL2 in knockout systems to confirm specificity .

Mechanistic validation:

  • Direct target identification: ChIP-seq or CUT&Tag approaches identify direct FOSL2 binding sites, distinguishing direct transcriptional targets from indirect effects.

  • Pathway perturbation: Manipulation of downstream mediators (like CCL28) can determine whether they are necessary for FOSL2-induced phenotypes.

  • Binding site verification: ChIP-qPCR and dual-luciferase reporter assays have demonstrated that FOSL2 directly binds and activates the CCL28 promoter, establishing causation rather than mere correlation .

Model systems:

  • In vivo xenograft models provide evidence for causality in a physiological context .

  • Using multiple cell types and model systems helps establish the generalizability of findings.

Research in pancreatic cancer employed these complementary approaches to establish that FOSL2 causally promotes cell proliferation, migration, invasion, and immune suppression, rather than merely correlating with these phenotypes .

What are the limitations of current FOSL2 research methodologies and how might they be addressed?

Current FOSL2 research faces several methodological limitations:

Cross-sectional vs. longitudinal approaches:
Many studies, including analyses of FOSL2 expression across aging, employ cross-sectional designs that may not capture individual changes over time .

Solutions:

  • Develop longitudinal study designs where feasible

  • Utilize paired samples from the same individuals when available

  • Apply mathematical modeling to infer temporal dynamics from cross-sectional data

Single-gene focus limitations:
Examining FOSL2 in isolation overlooks interactions with other AP-1 family members and cooperative transcription factors .

Solutions:

  • Expand studies to include multiple AP-1 family members simultaneously

  • Apply systems biology approaches to map interaction networks

  • Develop computational models accounting for combinatorial effects

Technical variability challenges:
Expression measurements are subject to variability that can introduce biases, particularly when comparing across experimental batches .

Solutions:

  • Implement robust normalization procedures

  • Include technical replicates and appropriate controls

  • Validate key findings using orthogonal methods

Translation between models and human disease:
Findings from model systems may not directly translate to human disease contexts due to species-specific differences in FOSL2 regulation .

Solutions:

  • Validate key findings across multiple model systems

  • Compare FOSL2 binding sites and target genes across species

  • Focus on conserved regulatory mechanisms

Addressing these limitations requires interdisciplinary approaches combining advanced experimental methods, computational modeling, and careful study design to fully elucidate FOSL2's complex roles in human biology and disease.

How should contradictory findings about FOSL2 function across different tissues and disease contexts be reconciled?

Contradictory findings regarding FOSL2 function reflect its context-dependent nature. Reconciling such discrepancies requires systematic approaches:

Context specificity analysis:
FOSL2 functions differently across cell types and biological contexts. For example, while FOSL2 promotes immune suppression in pancreatic cancer by activating CCL28 , it may have distinct functions in other cellular environments. Researchers should:

  • Define the cellular context of each study explicitly

  • Compare experimental conditions, particularly cell type, activation state, and microenvironmental factors

  • Consider species differences when comparing human and mouse studies

Temporal dynamics consideration:
FOSL2 activity varies across time points. Studies examining FOSL2 in Th17 cells noted the importance of timing, with reliable detection of relevant markers only after 72 hours of differentiation . Researchers should:

  • Compare timepoints used across studies

  • Consider acute versus chronic effects

  • Evaluate whether developmental stages influence results

Methodological comparison:
When conflicting results emerge, researchers should examine:

  • Knockdown/knockout strategies (acute vs. constitutive, partial vs. complete)

  • Overexpression systems (physiological vs. supraphysiological levels)

  • Detection methods (antibody specificity, RNA vs. protein measurements)

  • Data analysis pipelines (normalization methods, statistical approaches)

Integrated multi-omics approaches:
To resolve contradictions, researchers should:

  • Combine transcriptomic and proteomic data

  • Incorporate epigenomic data to understand context-specific accessibility

  • Validate key findings using orthogonal methods

When reporting results, researchers should clearly distinguish between correlative and causal evidence, explicitly state methodological limitations, and avoid causal language when only associative data is available.

What are the most promising therapeutic implications of targeting FOSL2 in human diseases?

The emerging understanding of FOSL2's role in disease pathogenesis reveals several promising therapeutic avenues:

Cancer immunotherapy approaches:
The discovery that FOSL2 promotes immunosuppression in pancreatic cancer by activating CCL28 and recruiting regulatory T cells suggests potential therapeutic strategies:

  • Targeting the KRAS-FOSL2-CCL28 axis could potentially restore anti-tumor immunity

  • Combining FOSL2 inhibition with existing immunotherapies might overcome resistance mechanisms

  • Monitoring FOSL2 expression could serve as a biomarker for predicting immunotherapy response

Age-related regenerative medicine:
The significant negative correlation between FOSL2 expression and age in mesenchymal stromal cells suggests therapeutic potential:

  • Modulation of FOSL2 might enhance the regenerative capacity of aging stem cells

  • FOSL2 expression profiles could help select more efficacious cells for therapeutic applications

  • Targeting FOSL2 might address the challenges posed by inherent variability in donor-derived cells' regenerative potential

Autoimmune disease interventions:
Given FOSL2's role in Th17 cell differentiation and function, targeting this pathway could offer therapeutic benefits:

  • Modulating FOSL2 activity might help regulate pathogenic Th17 responses in autoimmunity

  • The FOSL2 interactome provides multiple potential targets for intervention in Th17-mediated diseases

  • Understanding how FOSL2 coordinates with RNA-binding proteins could reveal novel therapeutic strategies

Future research should focus on developing specific inhibitors of FOSL2 activity or its critical downstream pathways, validating these approaches in preclinical models, and identifying biomarkers to select patients most likely to benefit from FOSL2-targeted therapies.

Product Science Overview

Gene and Protein Structure

The FOSL2 gene is located on chromosome 2 at the position 2p23.2 . The gene spans approximately 24,869 base pairs and is composed of several exons . The protein encoded by this gene has a molecular weight of approximately 35 kDa .

Function

FOSL2 plays a crucial role in regulating various cellular processes, including:

  • Cell Proliferation: FOSL2, as part of the AP-1 complex, regulates the expression of genes involved in cell cycle progression and proliferation .
  • Differentiation: It is involved in the differentiation of various cell types, including osteoclasts and keratinocytes .
  • Transformation: FOSL2 has been implicated in cellular transformation and oncogenesis .
Mechanism of Action

FOSL2 functions primarily as a transcription factor. It binds to specific DNA sequences in the promoter regions of target genes, regulating their transcription . The protein forms heterodimers with JUN family proteins, enhancing its DNA-binding affinity and specificity . This interaction is critical for the formation of the AP-1 transcription factor complex, which regulates the expression of genes involved in various cellular processes .

Clinical Significance

FOSL2 has been associated with several diseases and conditions, including:

  • Breast Cancer: Overexpression of FOSL2 has been linked to the progression and metastasis of breast cancer .
  • Pheochromocytoma: Altered expression of FOSL2 has been observed in pheochromocytoma, a type of adrenal gland tumor .
Research and Applications

Recombinant FOS-Like Antigen 2 (Human) is widely used in research to study its role in cellular processes and disease mechanisms. It is also utilized in drug discovery and development, particularly in identifying potential therapeutic targets for cancer and other diseases .

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.