ATF4 Human

Activating Transcription Factor-4 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Overview of ATF4 Human

ATF4 (Activating Transcription Factor 4) is a stress-responsive transcription factor encoded by the ATF4 gene in humans. It belongs to the basic leucine zipper (bZIP) superfamily and regulates adaptive cellular processes during metabolic, oxidative, and endoplasmic reticulum (ER) stress . ATF4 is ubiquitously expressed and controls genes involved in amino acid metabolism, redox homeostasis, autophagy, and apoptosis .

Gene and Protein Structure

  • Gene location: Chromosome 22q13.1

  • Protein domains:

    • Basic region: DNA-binding domain.

    • Leucine zipper: Facilitates dimerization with other bZIP proteins (e.g., C/EBPβ) .

  • Isoforms: Two transcript variants encode the same 351-amino-acid protein .

Regulatory Mechanisms

ATF4 activation occurs via:

  • Phosphorylation of eIF2α: Triggered by kinases (PERK, GCN2, PKR, HRI) under stress, enabling selective translation of ATF4 mRNA .

  • mTORC1 signaling: Promotes ATF4 translation independently of eIF2α phosphorylation to regulate anabolic processes .

  • Post-translational modifications: Phosphorylation, ubiquitination, and acetylation modulate its stability and activity .

Stress Adaptation

Stress TypeATF4-Mediated ResponseKey Targets
ER StressActivates unfolded protein response (UPR); regulates amino acid synthesis and redox genesCHOP, GADD34, SLC7A11
Oxidative StressEnhances glutathione synthesis via NADPH productionG6PDX, MTHFD2, PHGDH
Nutrient DeprivationInduces amino acid transporters and tRNA-charging enzymesASNS, SLC1A5, PSAT1

Disease Associations

  • Cancer: Promotes tumor survival by upregulating antioxidant pathways (e.g., SLC7A11 for cystine uptake) and conferring chemotherapy resistance .

  • Cardiovascular Disease: Protects against heart failure by reducing oxidative stress and apoptosis in cardiomyocytes .

  • Metabolic Disorders: Regulates hepatic cholesterol metabolism and mitigates hepatosteatosis .

ATF4 in Metabolic Regulation

  • mTORC1-ATF4 Axis: Drives protein synthesis and glutathione production by inducing tRNA-charging enzymes (e.g., MTHFD2) and the cystine transporter SLC7A11 .

  • Mitochondrial Function: Maintains NADPH/NADP+ balance via one-carbon metabolism and the pentose phosphate pathway .

ATF4 in Cellular Survival and Death

  • Pro-Survival: Activates autophagy and antioxidant genes (e.g., HMOX1) during nutrient deprivation .

  • Pro-Apoptotic: Collaborates with CHOP to induce GADD34 and BBC3/PUMA under unresolved ER stress .

Therapeutic Targets

  • Cancer: ATF4 inhibition sensitizes tumors to ferroptosis-inducing therapies (e.g., sorafenib in hepatocellular carcinoma) .

  • Cardiovascular Disease: ATF4 activation reduces oxidative damage in pressure-overloaded hearts .

  • Liver Disease: ATF4 maintains cholesterol homeostasis and prevents hepatocyte death during ER stress .

Genetic and Pharmacological Modulation

  • ATF4 Knockout Models: Cardiomyocyte-specific deletion exacerbates heart failure post-pressure overload .

  • Small-Molecule Activators: Compounds targeting eIF2α phosphorylation (e.g., ISRIB) modulate ATF4 activity .

Table 2: ATF4-Associated Pathways

PathwayRole of ATF4References
Integrated Stress ResponseCentral transcriptional effector of eIF2α signaling
Amino Acid MetabolismInduces asparagine synthetase (ASNS) and serine biosynthesis enzymes
Redox HomeostasisUpregulates glutathione synthesis via SLC7A11 and G6PDX

Product Specs

Introduction
Activating transcription factor 4 (ATF4) is a DNA-binding protein belonging to a family that includes AP-1 transcription factors, cAMP-response element binding proteins, and CREB-like proteins. It was first discovered as a DNA binding protein found in mammals that could bind to a tax-responsive enhancer element in the LTR of HTLV-1.
Description
Recombinant human ATF4 protein, produced in E. coli, is a single, non-glycosylated polypeptide chain of 510 amino acids (amino acids 1-351). It has a molecular weight of 56.6 kDa. For purification purposes, the ATF4 protein is tagged at the N-terminus with a 159 amino acid His-Calmodulin tag.
Physical Appearance
ATF4 is a clear, sterile solution.
Formulation
ATF4 protein solution (0.5 mg/ml) in 20 mM Tris-HCl buffer (pH 8.0), 1 mM DTT, and 10% glycerol.
Stability
For short-term storage (2-4 weeks), store at 4°C. For longer 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 determined by SDS-PAGE is greater than 90%.
Synonyms
Cyclic AMP-dependent transcription factor ATF-4, cAMP-dependent transcription factor, ATF-4, Activating transcription factor 4, Cyclic AMP-responsive element-binding protein 2, CREB-2, cAMP-responsive element-binding protein 2, DNA-binding protein, TAXREB67, Tax-responsive enhancer element-binding protein 67, TaxREB67, ATF4, CREB2, TXREB.
Source
Escherichia Coli.
Amino Acid Sequence
MHHHHHHMAD QLTEEQIAEF KEAFSLFDKD GDGTITTKEL GTVMRSLGQN PTEAELQDMI NEVDADGNGT IDFPEFLTMM ARKMKDTDSE EEIREAFRVF DKDGNGYISA AELRHVMTNL GEKLTDEEVD EMIREADIDG DGQVNYEEFV QMMTAKGSHM TEMSFLSSEV LVGDLMSPFD
QSGLGAEESL GLLDDYLEVA KHFKPHGFSS DKAKAGSSEW LAVDGLVSPS NNSKEDAFSG TDWMLEKMDL KEFDLDALLG IDDLETMPDD LLTTLDDTCD LFAPLVQETN KQPPQTVNPI GHLPESLTKP DQVAPFTFLQ PLPLSPGVLS STPDHSFSLE LGSEVDITEG DRKPDYTAYV
AMIPQCIKEE DTPSDNDSGI CMSPESYLGS PQHSPSTRGS PNRSLPSPGV LCGSARPKPY DPPGEKMVAA KVKGEKLDKK LKKMEQNKTA ATRYRQKKRA EQEALTGECK ELEKKNEALK ERADSLAKEI QYLKDLIEEV RKARGKKRVP.

Q&A

What is ATF4 and what is its primary function in human cells?

ATF4 is a multifunctional transcription regulatory protein belonging to the basic leucine zipper (bZIP) superfamily. Despite its name suggesting standalone transcriptional activation capabilities, ATF4 actually functions as one-half of heterodimeric transcription factors, partnering with other bZIP family members to regulate gene expression .

ATF4 coordinates cellular responses to various stress conditions and growth factors by regulating genes involved in amino acid transport and metabolism, redox balance, protein folding, and under severe stress conditions, apoptosis. It serves as a central node in the integrated stress response, allowing cells to adapt to challenging environmental conditions .

How is ATF4 expression regulated at the translational level?

  • Stress-induced eIF2α phosphorylation: The integrated stress response involves activation of one of four eIF2α kinases (GCN2, HRI, PERK, and PKR) in response to specific stressors. These kinases phosphorylate eIF2α, inhibiting eIF2B and allowing ribosomes to bypass inhibitory upstream open reading frames in the ATF4 mRNA .

  • Growth factor signaling: Anabolic hormones and growth factors like insulin, IGF-1, and TGFβ can increase ATF4 translation by activating mTORC1, which facilitates ribosomal bypass of inhibitory elements in the ATF4 mRNA through mechanisms distinct from eIF2α phosphorylation .

This translational control allows for rapid ATF4 induction in response to stress while simultaneously decreasing general protein synthesis to conserve resources.

What cellular stressors activate each of the eIF2α kinases that regulate ATF4?

ATF4 translation increases in response to various cellular stressors, each activating specific eIF2α kinases:

eIF2α KinaseActivating StressorsResult
GCN2Amino acid deprivation, UV radiationActivates amino acid response pathway
HRIOxidative stress, mitochondrial stress, heme deficiencyProtects against oxidative damage
PERKEndoplasmic reticulum stressTriggers unfolded protein response
PKRViral infections (double-stranded RNA)Initiates antiviral responses

All four kinases converge on eIF2α phosphorylation, which inhibits general translation while selectively increasing ATF4 translation .

How does ATF4 switch between promoting cell survival and inducing apoptosis?

The dual role of ATF4 in both promoting survival and inducing apoptosis depends on several factors:

  • Heterodimer formation: Different ATF4 binding partners influence target gene selection. Dimerization with C/EBPβ and C/EBPγ is associated with adaptive responses, while dimerization with CHOP has been linked to pro-apoptotic signaling .

  • Stress duration and intensity: Prolonged or severe stress shifts ATF4 function toward pro-apoptotic gene expression. Under persistent stress conditions, ATF4 can promote degenerative conditions such as skeletal muscle atrophy .

  • Post-translational modifications: Various PTMs may alter ATF4's function, DNA binding properties, and interaction partners, contributing to the switch between pro-survival and pro-apoptotic activities .

  • Cellular context: The existing metabolic state and signaling network of the cell influence how ATF4 activation affects cell fate. Cancer cells might be more resistant to pro-apoptotic ATF4 targets due to defects in downstream apoptotic signaling .

This complex regulatory network allows for context-specific responses to stress while creating challenges for predicting the outcome of ATF4 activation in different cellular contexts.

What genes and biological processes does ATF4 regulate?

ATF4, through heterodimer formation with various partners, regulates genes involved in numerous biological processes:

Biological ProcessTarget Gene CategoriesFunctional Outcome
Amino acid metabolismTransporters, aminoacyl-tRNA synthetasesIncreased amino acid availability
Redox homeostasisAntioxidant enzymes, glutathione synthesisProtection against oxidative stress
Protein foldingMolecular chaperones, heat shock proteinsEnhanced protein folding capacity
AutophagyAutophagy-related genesCellular component recycling
ApoptosisPro-apoptotic factorsProgrammed cell death (under severe stress)
Translation controlGADD34Feedback regulation of eIF2α phosphorylation

The specific genes regulated depend on cellular context, stress type and duration, and ATF4's heterodimeric partners .

How do ATF4 heterodimers form and how does this affect DNA binding specificity?

ATF4 functions exclusively through heterodimer formation with members of the ATF, FOS/JUN, and CCAAT enhancer-binding protein (C/EBP) bZIP transcription factor subfamilies. These heterodimers bind to DNA sequences called cAMP responsive elements (CREs) or C/EBP-ATF response elements (CAREs) .

Key aspects of ATF4 heterodimer formation:

  • Partner availability: ATF4 homodimers are unstable even when bound to DNA, making heterodimer formation essential for function .

  • DNA binding specificity: Different heterodimers have distinct preferences for slight variations in binding site sequences, affecting which genes are regulated.

  • Context-dependent partner selection: The composition of heterodimers varies with cell type and stress conditions, contributing to the versatility of ATF4 responses.

  • Functional consequences: ATF4-C/EBPβ and ATF4-C/EBPγ heterodimers have been associated with adaptive responses, while ATF4-CHOP heterodimers are traditionally linked to pro-apoptotic gene expression .

This heterodimer diversity adds flexibility to ATF4-mediated transcriptional responses, allowing cells to fine-tune their response to different stress conditions.

How is ATF4 dysregulation implicated in cancer biology?

ATF4 is frequently upregulated in cancer cells and plays complex roles in tumor development and progression:

Pro-tumor functions:

  • Facilitates adaptation to hypoxic tumor microenvironments

  • Promotes amino acid uptake and metabolism supporting cancer cell proliferation

  • Enhances glutathione synthesis to combat oxidative stress

  • Activates autophagy, which can support cancer cell survival under nutrient limitation

  • Intersects with mTOR signaling to coordinate anabolic processes

Potential anti-tumor functions:

  • Can promote apoptosis under severe stress conditions

  • May limit proliferation during acute stress

Cancer cells may exploit ATF4's pro-survival functions while developing resistance to its pro-apoptotic effects through alterations in downstream pathways. This makes ATF4 a potential therapeutic target, where strategies to push cancer cells toward ATF4-mediated apoptosis might be effective .

What role does ATF4 play in skeletal muscle atrophy and sarcopenia?

ATF4 has emerged as an important regulator in skeletal muscle homeostasis and pathology:

  • Muscle atrophy mechanisms: Sustained ATF4 activation can promote skeletal muscle atrophy. If a cellular stress continues unabated, ATF4 heterodimers can shift from adaptive to degenerative functions, contributing to conditions such as skeletal muscle atrophy .

  • Age-related muscle loss: ATF4 may contribute to sarcopenia through effects on muscle protein synthesis and breakdown balance.

  • Therapeutic targeting: Compounds like tomatidine and ursolic acid may affect ATF4 signaling in muscle, highlighting ATF4 as a potential therapeutic target for conditions involving skeletal muscle wasting .

The dual role of ATF4 in both adaptation and degeneration makes it a complex but important factor in muscle biology and pathology.

What methodological challenges exist in measuring ATF4 protein levels and activity?

Researchers face several technical challenges when investigating ATF4:

  • Protein detection difficulties:

    • ATF4 protein is typically expressed at low levels under basal conditions

    • The protein has a short half-life

    • Commercial antibodies vary in specificity and sensitivity

  • Translational regulation complexity:

    • mRNA measurements may not reflect protein abundance due to translational control

    • Specialized techniques such as polysome profiling are needed to assess translational efficiency

    • Reporter constructs containing ATF4 uORFs require careful design and interpretation

  • Heterodimer dynamics:

    • ATF4 heterodimers are unstable even when bound to DNA

    • The composition of heterodimers may change rapidly with cellular conditions

    • Detecting specific heterodimers in their native context is technically challenging

  • Activity assessment:

    • ATF4 binding to DNA does not necessarily indicate transcriptional activation

    • Post-translational modifications affect activity but are difficult to monitor comprehensively

    • Target gene expression is influenced by multiple factors beyond ATF4 binding

Addressing these challenges requires combining multiple complementary techniques and careful experimental design.

What experimental approaches are used to study ATF4 activation and function?

Researchers employ various experimental approaches to study ATF4:

  • Genetic models:

    • ATF4 knockout mice and cells (such as the Atf4 tm1Tow null mouse model)

    • Conditional knockouts for tissue-specific studies

    • CRISPR-Cas9 genome editing for precise manipulation

  • Stress induction models:

    • Thapsigargin or tunicamycin treatment for ER stress

    • Amino acid starvation media

    • Oxidative stress inducers

    • Hypoxia chambers

    • UV irradiation

  • Protein analysis:

    • Western blotting to detect ATF4 protein levels

    • Co-immunoprecipitation to identify interaction partners

    • Chromatin immunoprecipitation (ChIP) to map DNA binding sites

  • Gene expression analysis:

    • RNA-seq for genome-wide expression profiling (as used in the lens study of Atf4 null mice)

    • RT-qPCR for targeted analysis of ATF4 target genes

    • Reporter assays with ATF4-responsive promoters

  • Translational control assessment:

    • Polysome profiling to assess translational efficiency

    • Reporter constructs containing ATF4 uORFs

These approaches, often used in combination, allow for comprehensive investigation of ATF4 biology from molecular mechanisms to physiological impacts.

How do ATF4 and mTOR pathways interact to coordinate cellular metabolism?

ATF4 signaling and mTOR pathways intersect at multiple levels, creating a complex regulatory network:

  • mTOR activation of ATF4: The mTORC1 complex can increase ATF4 translation in response to growth factors like insulin and IGF-1, though through mechanisms distinct from eIF2α phosphorylation .

  • ATF4 enhancement of mTOR activity: ATF4 can enhance mTOR activity by:

    • Increasing amino acid availability through the upregulation of amino acid transporters

    • Inducing autophagy, which can generate amino acids

    • Upregulating genes involved in amino acid synthesis

  • Complementary roles in anabolism: In growth-promoting conditions, mTOR activates protein synthesis while ATF4 ensures sufficient amino acid supply for this increased synthetic demand.

  • Contextual interactions: During insulin/IGF-1 signaling, ATF4 heterodimers act in the setting of nutrient abundance and numerous other hormone-mediated events that promote anabolism and inhibit catabolism .

This crosstalk between ATF4 and mTOR pathways allows for coordinated responses to changing cellular conditions, balancing protein synthesis with amino acid availability.

How do post-translational modifications affect ATF4 function?

Post-translational modifications (PTMs) of ATF4 provide an additional layer of regulation that can influence its stability, localization, DNA binding, and interactions with other proteins:

Modification TypePotential SitesFunctional Effects
PhosphorylationMultiple residuesAlters stability, activity, partner binding
UbiquitinationLysine residuesControls protein degradation and turnover
SUMOylationPredictedMay affect localization and transcriptional activity
AcetylationPredictedCould influence DNA binding or protein interactions

According to research on human ATF4, evidence for these modifications comes from both mass spectrometry screening and targeted validation approaches .

These PTMs likely contribute to the context-dependent functions of ATF4, potentially influencing which target genes are activated under different conditions and possibly contributing to the switch between pro-survival and pro-apoptotic functions.

Product Science Overview

Structure and Function

ATF4 is a basic leucine zipper (bZIP) transcription factor that binds to specific DNA sequences to regulate the transcription of target genes . It is involved in the unfolded protein response (UPR), a cellular stress response related to the ER . Under stress conditions, ATF4 is upregulated and activates genes that help the cell adapt and survive .

Role in Cellular Stress

ATF4 is a key player in the cellular stress response. It helps cells cope with ER stress by regulating the expression of genes involved in protein folding, antioxidant responses, and amino acid metabolism . In addition, ATF4 is involved in the regulation of ferroptosis, a form of programmed cell death characterized by iron-dependent lipid peroxidation . The dual role of ATF4 in promoting cell survival and ferroptosis highlights its complex function in cellular stress responses .

Involvement in Diseases

ATF4 has been implicated in various diseases, including cancer, neurodegenerative disorders, and metabolic diseases . In cancer, ATF4 can either promote or inhibit tumor growth depending on the context . Its role in neurodegenerative diseases is linked to its function in managing oxidative stress and protein misfolding .

Research and Applications

Recent research has focused on understanding the genome-wide binding sites of ATF4 and its interaction with genetic variants associated with various traits and diseases . For example, studies have identified single-nucleotide polymorphisms (SNPs) that affect ATF4-mediated transcriptional activation, providing insights into the molecular mechanisms underlying these genetic associations .

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.