ATF1 Human

Activating Transcription Factor-1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Functions

ATF1 regulates diverse cellular processes:

Transcriptional Regulation

  • Modulates genes involved in proliferation, apoptosis, and stress responses via CRE binding .

  • Enhances cell transformation when phosphorylated, amplifying its transactivation activity .

Innate Immunity

  • Restricts HHV-6A (human herpesvirus 6A) replication by inducing interferon-β (IFN-β) expression. Knockout of ATF1 increases viral yields by 10–100-fold .

Developmental Biology

  • Acts as a gatekeeper for neuroectoderm (NE) differentiation in human embryonic stem cells (hESCs). ATF1 knockdown upregulates SOX2 and PAX6, accelerating NE lineage commitment .

Clinical Significance

ATF1 dysregulation is implicated in multiple pathologies:

Oncogenic Fusion Proteins

Fusion PartnerAssociated CancerMechanism
EWSR1Clear Cell Sarcoma (CCS)Drives tumorigenesis by hijacking enhancer regions and upregulating proliferation genes (e.g., MITF, DCT)
FUSAngiomatoid Fibrous HistiocytomaPromotes cell transformation via aberrant transcriptional programs

Key Findings in CCS

  • The EWSR1::ATF1 fusion protein binds distal enhancer regions, altering chromatin conformation and activating oncogenic pathways .

  • ATF1 knockdown in CCS cells restores Schwann cell/melanocyte differentiation markers (e.g., TYR, TPPP3) .

Neuroinflammatory Diseases

  • HHV-6A leverages ATF1-CREB1 signaling to evade immune detection, linking ATF1 to viral persistence in neuroinflammatory conditions .

Transcriptomic Insights

  • In hESCs, ATF1 suppresses NE differentiation by repressing SOX2. CRISPR/Cas9 knockout of ATF1 increases SOX2 expression by >3-fold .

  • ATF1 deficiency downregulates innate immune sensors (e.g., TLR3, RIG-I), reducing IFN-β production during viral infection .

Therapeutic Targets

  • Targeting EWSR1::ATF1 binding to enhancers (e.g., with epigenetic modifiers) induces senescence in CCS cells .

  • IFN-β supplementation reverses HHV-6A replication in ATF1-knockout models, suggesting immunomodulatory strategies .

Interaction Network

ATF1 collaborates with:

  • BRCA1: Modulates DNA repair pathways .

  • EWS: Forms oncogenic chimeras in sarcomas .

  • Casein Kinase 2 (CK2): Regulates post-translational modifications .

Future Directions

  • Elucidate ATF1’s role in balancing pluripotency and differentiation in stem cells.

  • Develop inhibitors targeting ATF1 fusion proteins in CCS and other malignancies.

Product Specs

Introduction
ATF1, a cyclic-AMP dependent transcription factor, is found in many cell types and can interact with CREB to form a dimer. MSK1 and MSK2 protein kinases are crucial for the stress-induced phosphorylation of transcription factors CREB and ATF1 in primary embryonic fibroblasts. Epidermal growth factor stimulation of c-jun expression requires ATF1 and MEF2 binding sites in the c-jun promoter.
Description
Recombinant human ATF1, produced in E. coli, is a single, non-glycosylated polypeptide chain comprising 295 amino acids (residues 1-271). It has a molecular weight of 31.8 kDa. ATF1 is expressed with a 24 amino acid His-tag at the N-terminus and is purified using proprietary chromatographic methods.
Physical Appearance
Clear, colorless solution, sterile-filtered.
Formulation
The ATF1 solution is provided at a concentration of 0.25 mg/ml in a buffer consisting of 20 mM Tris-HCl (pH 8.0), 200 mM NaCl, 5 mM DTT, 2 mM EDTA, and 50% glycerol.
Stability
Store at 4°C if the entire vial will be used within 2-4 weeks. For extended storage, freeze at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
Purity greater than 85% as determined by SDS-PAGE analysis.
Synonyms
Activating transcription factor 1, cyclic AMP-dependent transcription factor ATF-1, Protein TREB36, EWS-ATF1, FUS/ATF-1.
Source
E.coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSHMEDSHK STTSETAPQP GSAVQGAHIS HIAQQVSSLS ESEESQDSSD SIGSSQKAHG ILARRPSYRK ILKDLSSEDT RGRKGDGENS GVSAAVTSMS VPTPIYQTSS GQYIAIAPNG ALQLASPGTD GVQGLQTLTM TNSGSTQQGT TILQYAQTSD GQQILVPSNQ VVVQTASGDM QTYQIRTTPS ATSLPQTVVM TSPVTLTSQT TKTDDPQLKR EIRLMKNREA ARECRRKKKE YVKCLENRVA VLENQNKTLI EELKTLKDLY SNKSV

Q&A

What is ATF1 and what is its primary function in human cells?

ATF1 (Activating Transcription Factor 1) is a basic region-leucine zipper transcription factor that belongs to the cAMP response element-binding protein (CREB) family . It functions as a transcriptional regulator that can both activate and repress gene expression depending on cellular context. In human embryonic stem cells (hESCs), ATF1 acts as a pluripotent regulator, specifically functioning as a negative transcriptional regulator of SOX2 expression . Research has demonstrated that ATF1 serves as a gatekeeper for neural lineage specification, maintaining the pluripotent state by preventing premature neuroectoderm differentiation .

What experimental methods are most effective for studying ATF1 expression in human samples?

Several methodological approaches have proven effective for studying ATF1 expression:

MethodApplicationAdvantagesLimitations
RT-qPCRmRNA expression quantificationHigh sensitivity, quantitativeDoes not measure protein levels
Western blottingProtein expression analysisDistinguishes between phosphorylated and non-phosphorylated formsSemi-quantitative
ImmunohistochemistryTissue localizationProvides spatial informationLimited quantification
RNA-seqTranscriptome-wide effectsGlobal gene expression analysisRequires bioinformatic expertise
ChIP-seqGenome-wide binding sitesIdentifies direct targetsLabor-intensive, requires optimization

When analyzing ATF1 activity, researchers should consider both total ATF1 levels and its phosphorylation status, as functional effects often depend on post-translational modifications .

How does ATF1 expression vary across different human tissues and cell types?

ATF1 expression patterns vary significantly across human tissues and developmental stages. In early embryonic development, active ATF1 accumulates in the 2-cell embryo stage . In human embryonic stem cells, ATF1 maintains pluripotency and is spontaneously down-regulated after 1-3 days of neural induction . Tissue-specific expression patterns reflect ATF1's diverse roles in different cellular contexts, including immune cells where it contributes to antiviral responses .

What is the role of ATF1 in maintaining human embryonic stem cell pluripotency?

ATF1 functions as a crucial pluripotent regulator in human embryonic stem cells (hESCs). Research has revealed that it acts as a "gatekeeper" for neural lineage specification by:

  • Negatively regulating SOX2 expression

  • Preventing premature differentiation toward neuroectoderm lineage

  • Maintaining the pluripotent state through transcriptional repression mechanisms

Down-regulation of ATF1 significantly up-regulates neuroectoderm genes but not mesoderm, endoderm, or trophectoderm genes, indicating its specific role in preventing neural differentiation . Luciferase reporter assays have confirmed that ATF1 acts as a negative transcriptional regulator of the SOX2 gene, one of the master regulators of neural development .

How does ATF1 knockdown affect neural differentiation pathways in human cells?

Knockdown or knockout of ATF1 using shRNA, siRNA, or CRISPR/Cas9 techniques leads to several significant changes in neural differentiation pathways:

  • Up-regulation of neuroectoderm markers, particularly SOX2 and PAX6, even under undifferentiated conditions

  • Accelerated neural differentiation when cells are placed in neural induction conditions

  • Enhanced expression of neural progenitor markers during the differentiation process

This indicates that ATF1 functions as a barrier to neural differentiation, and its removal is sufficient to initiate neuroectoderm specification pathways. Conversely, overexpression of ATF1 suppresses neural differentiation even under conditions that normally promote it .

What techniques are most reliable for ATF1 knockdown or knockout studies in human stem cells?

When studying ATF1 function through loss-of-function approaches, researchers have several methodological options with different advantages:

TechniqueEfficiencyAdvantagesConsiderations
shRNAModerateStable knockdown, titratablePotential off-target effects
siRNAHigh, transientRapid effect, high transfection efficiencyShort duration of effect
CRISPR/Cas9Complete knockoutPermanent modification, no residual proteinClonal selection required

For hESC experiments specifically, studies have successfully used:

  • Short hairpin RNA (shRNA) targeting ATF1 with the sequence found in TRCN0000273833

  • CRISPR/Cas9 with sgRNA-Cas9-2A-PAC plasmid delivery using TransIT-LT1 transfection reagent

  • Flow cytometry sorting of GFP-positive cells for isolation of edited cells

For effective CRISPR/Cas9 knockout in hESCs, dissociation of cells with Accutase and treatment with 10 μM Rho-associated protein kinase inhibitor Y-27632 significantly improves cell survival during the editing process .

How does ATF1 contribute to antiviral immunity in human cells?

ATF1 plays a significant role in antiviral immune responses, particularly against herpesviruses such as Human Herpesvirus 6A (HHV-6A). Research has demonstrated that:

  • HHV-6A infection enhances phosphorylation of ATF1

  • ATF1 restricts HHV-6A replication through induction of beta interferon (IFN-β)

  • Knockout of ATF1 significantly enhances viral gene expression and replication

Transcriptome sequencing (RNA-seq) analysis revealed that ATF1 knockout leads to downregulation of innate immune system sensors and reduced expression of IFN-β and IFN-regulated genes during HHV-6A infection . Importantly, treatment with exogenous IFN-β can rescue the enhanced viral replication in ATF1-knockout cells, confirming that ATF1 restricts viral replication primarily through the IFN pathway .

What is the relationship between AMPK signaling and ATF1 activation in human macrophages?

In human macrophages, ATF1 is activated through the 5'-AMP-activated protein kinase (AMPK) pathway in response to heme exposure . This activation represents an important mechanism in cellular responses to intraplaque hemorrhage in atherosclerotic lesions. The activation sequence involves:

  • Heme exposure triggers AMPK activation in macrophages

  • AMPK activation leads to ATF1 phosphorylation and activation

  • Activated ATF1 initiates specific transcriptional programs in these cells

This signaling cascade appears to generate a distinct adaptive macrophage state (Mhem) that plays a role in the cellular response to hemorrhagic conditions within atherosclerotic plaques .

What experimental considerations are important when studying ATF1 phosphorylation?

When investigating ATF1 phosphorylation, researchers should consider:

  • Antibody selection: Use antibodies that specifically recognize phosphorylated ATF1 at key regulatory sites

  • Preservation of phosphorylation status: Include phosphatase inhibitors in all lysis buffers

  • Positive controls: Include samples treated with known activators of ATF1 phosphorylation (e.g., forskolin for cAMP pathway activation)

  • Kinase inhibitor studies: Use specific inhibitors to identify the kinases responsible for ATF1 phosphorylation

  • Time-course experiments: Monitor the dynamics of phosphorylation/dephosphorylation

Research has shown that both CREB1 and ATF1 phosphorylation increases during HHV-6A infection , suggesting viral triggers for this modification. Similarly, heme exposure triggers ATF1 phosphorylation via AMPK in macrophages , demonstrating the diverse stimuli that can activate this transcription factor.

How can ChIP-seq be optimized for studying ATF1 binding sites in human cells?

Chromatin Immunoprecipitation followed by sequencing (ChIP-seq) is a powerful technique for identifying genome-wide ATF1 binding sites. For optimal results:

Optimization StepRecommendationRationale
Crosslinking1% formaldehyde for 10 minutesBalances efficiency with specificity
SonicationOptimize to achieve 200-500bp fragmentsIdeal fragment size for sequencing
Antibody selectionValidate with known ATF1 targetsEnsures specificity of immunoprecipitation
Input controlsUse matched input chromatinCritical for accurate peak calling
Peak callingUse algorithms that account for ATF1's binding characteristicsImproves detection of genuine binding sites
Motif analysisInclude analysis of cAMP response elements (CREs)ATF1 typically binds to CRE motifs

Since ATF1 has been shown to act as a negative transcriptional regulator of SOX2 , ChIP-seq can be particularly valuable for identifying direct targets and distinguishing between direct transcriptional regulation and secondary effects.

What are the best approaches for studying ATF1 function in neural differentiation models?

When investigating ATF1's role in neural differentiation, several approaches have proven effective:

  • Staged differentiation protocols: The established protocol includes:

    • Treatment with dorsomorphin (2 μM) and A-83-01 (2 μM) in embryoid body medium

    • Reattachment on Matrigel-coated plates with neural progenitor cell medium

    • Supplementation with cyclopamine (2 μM) and heparin (2 μg/ml)

  • Functional validation of neural identity:

    • Electrophysiological recordings using whole-cell patch-clamp techniques

    • Membrane potentials held at −70 mV by voltage clamp

    • Analysis of action potentials and currents using specialized software

  • Comparative analysis:

    • Parallel differentiation of control and ATF1-knockdown cells

    • Temporal analysis of neural marker expression

    • Assessment of functional neuronal properties

These approaches provide complementary data on both molecular and functional aspects of neural differentiation as influenced by ATF1.

What are the current challenges in integrating multi-omics data for comprehensive ATF1 function analysis?

Researchers face several challenges when integrating multi-omics data to understand ATF1 function:

  • Data integration complexity: Combining ChIP-seq, RNA-seq, and proteomics data requires sophisticated computational approaches

  • Temporal dynamics: ATF1 function changes during developmental processes, requiring time-series analysis

  • Cell type specificity: ATF1 may have different targets and functions in different cell types

  • Contextual interpretation: The same ATF1 binding event may have different outcomes depending on cofactors present

  • Technical biases: Different omics platforms have inherent biases that must be accounted for

Current best practices include using matched samples for different omics analyses, employing integrative computational frameworks, and validating key findings with targeted experiments such as reporter assays, which have been successfully used to confirm ATF1's role as a repressor of SOX2 expression .

What are promising research areas for understanding ATF1's broader role in human development?

Several promising research directions for expanding our understanding of ATF1 include:

  • Developmental stage-specific functions: Investigating ATF1's changing roles across different stages of human development, from early embryogenesis through organogenesis

  • Cell type-specific regulatory networks: Mapping ATF1's interaction partners and target genes in different cell lineages

  • Single-cell analyses: Using single-cell transcriptomics and epigenomics to capture heterogeneity in ATF1 function

  • Mechanistic studies of dual activator/repressor function: Understanding how ATF1 can both activate and repress gene expression in different contexts

  • Integration with developmental signaling pathways: Exploring ATF1's relationship with key developmental pathways such as BMP, Wnt, and Notch signaling

These approaches would build upon the finding that ATF1 serves as a gatekeeper for neural lineage specification in hESCs and could reveal additional regulatory roles in other developmental processes.

How might ATF1-targeted therapies be developed for viral infections or neurological disorders?

Based on ATF1's roles in viral immunity and neural development, potential therapeutic approaches might include:

  • Antiviral strategies:

    • Enhancing ATF1 activity to boost IFN-β responses against herpesviruses

    • Developing small molecules that mimic ATF1's antiviral effects

    • Targeting viral mechanisms that may inhibit ATF1 function

  • Neurological applications:

    • Modulating ATF1 to enhance neural differentiation for cell replacement therapies

    • Targeting ATF1 in neural stem cell populations to promote regeneration

    • Investigating ATF1's role in neurological disorders with developmental origins

Development of such therapies would require detailed understanding of ATF1's tissue-specific functions and careful assessment of potential off-target effects, given ATF1's diverse roles across different cellular contexts.

Product Science Overview

Structure and Expression

ATF-1 is a protein coding gene that encodes a transcription factor. The human recombinant form of ATF-1 is produced in E. coli and is a single, non-glycosylated polypeptide chain containing 295 amino acids, with a molecular mass of approximately 31.8 kDa . The protein is fused to a 24 amino acid His-tag at the N-terminus and is purified using proprietary chromatographic techniques .

Functional Role

ATF-1 influences cellular physiological processes by regulating the expression of genes related to growth, survival, and other cellular activities . It is particularly significant in the context of atherosclerosis, where it mediates the induction of heme oxygenase-1 (HO-1) by heme and drives macrophage adaptation to intraplaque hemorrhage . This process is crucial for protecting macrophages from foam cell formation and oxidative stress, which are key factors in the progression of atherosclerosis .

Applications and Storage

The human recombinant ATF-1 is used in various research applications, including studies on gene regulation and cellular responses to stress. It is typically stored at -20°C for long-term storage, with the recommendation to add a carrier protein to prevent multiple freeze-thaw cycles .

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.