ATF3 (Activating Transcription Factor 3) is a stress-responsive transcription factor encoded by the ATF3 gene in humans. It belongs to the ATF/CREB protein family and regulates gene expression by binding to cyclic AMP response elements (CREs) in target promoters . ATF3 plays dual roles as both a transcriptional activator and repressor, depending on cellular context and dimerization partners . Its expression is induced by diverse stressors, including cytokines, DNA damage, and metabolic disturbances .
Isoforms:
Property | Details |
---|---|
Molecular weight | 22 kDa |
Key domains | bZIP, transcriptional activation domain |
Dimerization partners | JUN, JUND, SMAD3, p53 |
Post-translational modifications | Phosphorylation, ubiquitination |
Glucose homeostasis:
Macrophages:
T cells:
Cancer Type | ATF3 Role | Mechanism |
---|---|---|
Breast | Stromal ATF3 correlates with metastasis | Enhances myeloid cell-mediated ECM remodeling |
Liver | Promotes fibrosis | Activates SMAD3 and lnc-SCARNA10/TGF-β pathway |
Prostate | Tumor suppressor | Androgen-dependent expression inhibits growth |
Liver fibrosis: ATF3 overexpression in hepatic stellate cells drives ECM deposition via SMAD3 recruitment .
Diabetes: ATF3 knockout mice exhibit improved glucose tolerance but reduced β-cell function .
Cancer prognosis:
ATF3 inhibition reduces HSC activation in liver fibrosis models .
ATF3 agonists show potential in enhancing β-cell survival under oxidative stress .
Data from the Human Protein Atlas :
Tissue | Expression Level (nTPM) |
---|---|
Liver | High |
Lung | Moderate |
Adipose tissue | Low |
Cerebral cortex | Not detected |
ATF3-SMAD3 axis: Identified as a master regulator of TGF-β-driven fibrogenesis in liver and lung .
Immune modulation: ATF3 deficiency exacerbates LPS-induced lung injury but improves antiviral responses .
Ferroptosis: ATF3 suppresses lipid peroxidation in hepatocytes via GPX4 upregulation .
ATF3 is a member of the basic leucine zipper (bZip) family of transcription factors that functions primarily as a stress-inducible gene. Evolutionarily, ATF3 diverged relatively late in history, likely evolving from a gene duplication of FOS that occurred before the cnidarian-bilaterian divergence . As a conserved transcription factor across vertebrate species, ATF3 serves as an adaptive response element that can both activate and repress gene expression depending on cellular context and binding partners .
In normal, unstressed human tissues, ATF3 expression is typically maintained at very low or nearly undetectable levels . Its expression is tightly regulated and rapidly induced by various stress signals, including many that trigger the unfolded protein response (UPR). The adaptive response network involving ATF3 is activated by cellular stressors across multiple tissue types including liver, heart, kidney, and nervous system tissues . The regulatory mechanisms controlling ATF3 expression involve complex signaling cascades that respond to both intrinsic and extrinsic cellular stressors .
ATF3 is intricately linked to the unfolded protein response pathway, serving as both a downstream target and a modulator of UPR-associated gene expression . When cells experience ER stress, the UPR is activated, triggering signaling cascades that induce ATF3 expression. Once expressed, ATF3 can regulate genes involved in protein folding, degradation, and cellular adaptation to stress, effectively functioning as part of a feedback mechanism within the UPR network . Methodologically, researchers studying this relationship often employ ER stress inducers such as tunicamycin or thapsigargin to assess ATF3's role in UPR dynamics .
Immunohistochemical detection of ATF3 in human samples requires careful optimization due to the generally low expression levels and the sensitivity of results to fixation conditions. A successful protocol typically includes:
Tissue preparation: Proper fixation in 10% neutral buffered formalin for 24–48 hours
Antigen retrieval: Heat-induced epitope retrieval using citrate buffer (pH 6.0)
Blocking: 5-10% normal serum to minimize non-specific binding
Primary antibody: Dilution optimization is critical (typically 1:50 to 1:200)
Detection system: Amplification systems like avidin-biotin complex (ABC) or polymer-based detection systems
Researchers should note that ATF3 immunostaining is particularly sensitive to fixation time and methods, making pilot experiments essential to establish optimal conditions for specific tissue types .
When working with low-abundance ATF3 expression, researchers should consider:
Signal amplification: Employ tyramide signal amplification (TSA) or other amplification systems
Extended antibody incubation: Overnight primary antibody incubation at 4°C
Optimized antigen retrieval: Test multiple buffers and incubation times
Reduced background: Use specialized blocking reagents containing both proteins and detergents
Fresh tissue sections: When possible, use freshly cut sections from paraffin blocks
Additionally, comparing multiple antibodies from different manufacturers can help identify the optimal reagent for specific applications, as antibody sensitivity varies considerably across commercial sources .
Robust ATF3 research requires multiple experimental controls:
Positive tissue controls: Samples known to express ATF3 (e.g., stressed tissues)
Negative tissue controls: Samples known to lack ATF3 expression
Antibody controls: Primary antibody omission and isotype controls
Peptide competition: Pre-absorption of antibody with immunizing peptide
Alternative detection methods: Validation with RT-qPCR or Western blotting
For knockout validation experiments, tissues from ATF3 knockout animals or CRISPR-edited cell lines should be included whenever possible to confirm antibody specificity .
After nervous system injury, ATF3 expression undergoes dramatic temporal and spatial changes:
Species | Tissue | Baseline ATF3 | Post-Injury Response | Time Course | Cell Types |
---|---|---|---|---|---|
Zebrafish | Spinal cord | Nearly undetectable | Highly induced | 4 hours to 11 days | Motor neurons, axonal profiles |
Lamprey | Spinal cord | Nearly undetectable | Highly induced | Extended period | Motor neurons, axonal profiles |
Rodents | PNS (sciatic nerve) | Low | Highly induced | Hours to weeks | DRG neurons |
Rodents | CNS (spinal cord) | Low | Limited induction | Minimal | Rare neuronal populations |
In species with high regenerative capacity such as zebrafish and lamprey, ATF3 is among the most highly induced regeneration-associated genes following spinal cord injury. In contrast, the mammalian central nervous system shows limited ATF3 induction, correlating with poor regenerative outcomes .
Multiple lines of evidence support ATF3's role as a pro-regenerative factor:
Expression pattern: ATF3 is consistently upregulated in regeneration-competent neurons across vertebrate species
Loss-of-function studies: Genetic deletion or knockdown of ATF3 reduces axon regeneration in mouse and zebrafish models
Gain-of-function studies: Inducing ATF3 expression promotes axon sprouting, regrowth, or regeneration
Molecular targets: ATF3 regulates genes involved in axonal growth, including galanin and GRP
Temporal correlation: ATF3 expression timing coincides with the initiation of regenerative responses
The experimental approach for studying ATF3's pro-regenerative functions typically involves comparing high-regeneration model systems (zebrafish, lamprey, peripheral nerves) with low-regeneration systems (mammalian CNS) .
Significant differences in ATF3 expression exist across species with different regenerative capabilities:
Species | CNS Regenerative Capacity | ATF3 Expression Pattern | Notable Features |
---|---|---|---|
Zebrafish | High | Rapid, robust induction | Among most highly induced RAGs; peaks within hours |
Lamprey | High | Rapid, robust induction | Most highly induced RAG in both spinal cord and brain |
Rodents (PNS) | High | Robust induction | Gradual decrease over regeneration period |
Rodents (CNS) | Limited | Minimal induction | Does not readily occur around injury site |
These differences in ATF3 expression patterns suggest that robust early ATF3 induction may be a critical component of successful neural regeneration programs. Methodologically, comparative transcriptomics across these species provides valuable insights into conserved regenerative mechanisms .
ATF3 serves as a central node in cellular stress response networks through several mechanisms:
Transcriptional regulation: ATF3 can both activate and repress gene expression depending on context
Protein interactions: ATF3 interacts with multiple transcription factors including c-Jun and JunD
Integration of signals: ATF3 responds to diverse stress signals, integrating multiple cellular pathways
Temporal dynamics: ATF3's rapid induction allows for timely cellular adaptations
Cell-type specificity: Distinct functions in different cell populations adapt responses to tissue context
As a methodological approach, studying ATF3's role as a network hub typically involves identifying its binding partners through co-immunoprecipitation, identifying its genomic binding sites via ChIP-seq, and characterizing downstream gene expression changes through RNA-seq .
ATF3 regulates various downstream targets involved in cellular adaptation to stress:
Neuropeptides: Galanin and GRP, which modulate neuronal signaling and inflammation
Inflammatory mediators: ATF3 can suppress inflammatory responses in multiple contexts
Cell survival factors: ATF3 regulates genes involved in apoptosis and cell survival
Growth-associated proteins: Various factors promoting axonal growth and regeneration
Research methods to identify ATF3 targets include promoter analysis, ChIP-seq approaches, and gene expression profiling following ATF3 manipulation. The diverse binding partners of ATF3 contribute to its context-dependent effects on different target genes .
Based on ATF3's pro-regenerative properties, several therapeutic approaches may be developed:
Gene therapy: Viral vector-mediated delivery of ATF3 to injured neurons
Pre-formed protein complexes: Delivery of pre-dimerized c-Jun-ATF3 complexes
Small molecule modulators: Compounds that enhance endogenous ATF3 expression
Combination approaches: Targeting ATF3 alongside other regeneration-associated pathways (PTEN/mTOR, cAMP, KLFs)
Cell-specific targeting: Neuronal-specific delivery systems to avoid off-target effects
Important methodological considerations include determining optimal timing of intervention, delivery methods, and potential off-target effects given ATF3's roles in multiple tissues and cellular processes .
ATF3 expression in patient samples may serve as a biomarker for various conditions:
Tissue stress: Indicator of cellular stress responses in affected tissues
Regenerative potential: Possible predictor of regenerative outcomes following injury
Disease progression: Potential correlation with disease severity or progression
Treatment response: Possible indicator of cellular adaptation to therapeutic interventions
Methodologically, researchers should employ standardized IHC protocols on patient samples, with careful consideration of tissue fixation variability. Quantitative assessment of ATF3 expression and correlation with clinical parameters requires consistent scoring methods and appropriate statistical analysis .
Several challenges must be addressed when translating ATF3 research to human applications:
Species differences: Variations in ATF3 function and regulation between model organisms and humans
Cell-type specificity: Different effects in various cell populations requiring targeted approaches
Temporal considerations: Optimal timing for intervention may differ between models and humans
Off-target effects: ATF3's multiple roles in various tissues may lead to unintended consequences
Technical limitations: Detection challenges in human samples due to post-mortem changes and fixation variables
Methodological approaches to overcome these challenges include using humanized animal models, human-derived organoids or iPSC models, and careful validation in human tissue samples when available .
ATF3 is encoded by the ATF3 gene, which consists of four exons that encode a 181-amino acid protein with a molecular weight of approximately 22 kDa . The protein contains a basic leucine zipper (bZIP) domain, which allows it to bind DNA and interact with other proteins. ATF3 can function as both a transcriptional activator and repressor, depending on the context and the specific genes it regulates .
ATF3 is a stress-induced transcription factor that is rapidly upregulated in response to various stress signals, including endoplasmic reticulum (ER) stress, cytokines, chemokines, and lipopolysaccharides (LPS) . It acts as a hub in the cellular adaptive-response network, modulating metabolism, immunity, and oncogenesis .
ATF3 has been implicated in several human diseases, including cancer, atherosclerosis, infections, and hypospadias . Dysregulation of ATF3 can lead to altered gene expression profiles, contributing to the development and progression of these diseases . In cancer, ATF3 has been shown to regulate the expression of genes involved in cell proliferation, apoptosis, and metastasis .
ATF3 binds to specific DNA sequences in the promoters of target genes, often in collaboration with other transcription factors and co-activators . For example, ATF3 can co-localize with the major stress responder p53 at genomic sites, thereby collaborating with p53 to regulate p53 target gene expression upon DNA damage . Additionally, ATF3 can recruit chromatin-modifying enzymes to convert chromatin to a state permissive for transcription .
Recombinant ATF3 is used in research to study its role in various cellular processes and diseases. By using human recombinant ATF3, researchers can investigate the specific functions and regulatory mechanisms of this transcription factor in a controlled environment. This can lead to a better understanding of its role in disease and potentially identify new therapeutic targets.