Activating transcription factor 5 (ATF5) is a transcription factor that modulates gene expression by binding to various DNA regulatory elements. These include the cAMP response element (CRE) (consensus: 5'-GTGACGT[AC][AG]-3'), the ATF5-specific response element (ARE) (consensus: 5'-C[CT]TCT[CT]CCTT[AT]-3'), and the amino acid response element (AARE), found in numerous viral and cellular promoters. ATF5 plays a crucial role, often in a cell-type-specific manner, in regulating cell survival, proliferation, and differentiation. Its transcriptional activity is positively influenced by CCND3 and slightly inhibited by CDK4.
ATF5 is a significant regulator of cerebral cortex development, maintaining proliferation and preventing neuronal differentiation in cortical neuroprogenitor cells. Downregulation of ATF5 is essential for these cells to exit the cell cycle and differentiate. It also participates in the Hedgehog (SHH) signaling pathway, promoting cerebellar granule neuron progenitor cell proliferation. ATF5 is critical for the survival of mature olfactory sensory neurons (OSNs) and regulates the expression of OSN-specific genes. It may also be involved in osteogenic differentiation. Furthermore, ATF5 promotes cell proliferation and survival by synergistically inducing EGR1 expression with ELK1. Following acetylation by EP300, ATF5 binds to ARE sequences on target gene promoters, such as those of BCL2 and EGR1, exerting an anti-apoptotic effect through BCL2 regulation (cell-type dependent). ATF5 also cooperates with NR1I3/CAR in activating CYP2B6 transcription in the liver. In hepatic cells, it represses CRE-dependent transcription and inhibits proliferation by arresting the cell cycle at the G2/M phase. It may also function as a negative regulator of the IL1B signaling pathway in the liver. Upon IL1B stimulation, ATF5 collaborates with NLK to activate the transactivation activity of C/EBP subfamily members. Beyond its role as a transcription factor, ATF5 acts as a cofactor for CEBPB, activating CEBPA and promoting adipocyte differentiation. Finally, ATF5 regulates centrosome dynamics in a cell-cycle and centriole-age-dependent manner, forming a 9-foci symmetrical ring scaffold around the mother centriole to control centrosome function and the interaction between centrioles and pericentriolar material.
ATF5 (Activating Transcription Factor 5) is a 30.7 kDa protein belonging to the ATF/CREB transcription factor family. It functions critically in cellular stress responses, particularly during hypoxia and nutrient deprivation . The protein is characterized by a conserved C-terminal leucine zipper domain that facilitates dimerization and DNA binding, while its N-terminal domains exhibit significant variability allowing for diverse regulatory functions .
ATF5 localizes in both cytoplasm and nucleus, functioning as a dimer to bind DNA. It interacts with several proteins including CCND3 and PTP4A1, which are involved in cell cycle regulation and signal transduction pathways . One of its key roles is maintaining mitochondrial homeostasis and regulating mitochondrial quality control, particularly in response to exercise and cellular stress . ATF5 has been implicated in cancer pathogenesis, with altered expression observed in glioblastoma, breast cancer, and pancreatic carcinoma .
When selecting an ATF5 antibody, consider these critical factors:
Species reactivity: Determine whether the antibody recognizes ATF5 in your species of interest. Many commercially available antibodies detect human, mouse, and rat ATF5, but cross-reactivity varies significantly .
Application compatibility: Verify the antibody has been validated for your specific application:
Western blotting (WB)
Immunoprecipitation (IP)
Immunofluorescence (IF)
Immunohistochemistry (IHC)
ELISA
Monoclonal antibodies (e.g., E-10 clone) offer consistency across experiments but may recognize only specific epitopes
Polyclonal antibodies provide broader epitope recognition but potentially more background
Epitope location: Consider whether your experimental conditions might mask or modify the epitope. For studies investigating post-translational modifications like acetylation at K29, ensure the epitope doesn't include or isn't affected by this site .
Validation data: Review immunoblots, IHC images, and immunofluorescence data from manufacturers to assess specificity and expected banding patterns .
For successful Western blot detection of ATF5, consider the following protocol optimizations:
Use RIPA buffer supplemented with protease inhibitors
Include phosphatase inhibitors if studying phosphorylation states
For detecting acetylated ATF5, add deacetylase inhibitors like trichostatin A (TSA)
Load 20-40 μg of total protein per lane
Use 10-12% polyacrylamide gels for optimal separation
Expected MW: ~30.7 kDa, though post-translational modifications may alter migration
Semi-dry or wet transfer at 100V for 60-90 minutes
PVDF membranes generally yield better results than nitrocellulose for ATF5
Block with 5% non-fat milk or BSA in TBST
Primary antibody dilutions typically range from 1:500 to 1:2000
Incubate overnight at 4°C for optimal results
Include positive controls (cells known to express ATF5)
Use ATF5 knockdown samples as negative controls
For acetylation studies, compare samples with and without deacetylase inhibitors
HRP-conjugated secondary antibodies with enhanced chemiluminescence detection work well
For quantitative analysis, consider fluorescent secondary antibodies
ATF5 exhibits both nuclear and cytoplasmic localization, making correct fixation and permeabilization critical:
4% paraformaldehyde (10-15 minutes at room temperature) preserves morphology
Methanol fixation (-20°C for 10 minutes) can improve nuclear epitope accessibility
0.1-0.3% Triton X-100 for PFA-fixed cells (10 minutes)
Avoid excessive permeabilization which can affect nuclear membrane integrity
Block with 5-10% normal serum from secondary antibody host species
Include 0.1% BSA to reduce non-specific binding
Primary antibody dilutions typically 1:50-1:200 for immunofluorescence
Incubate overnight at 4°C in a humidified chamber
DAPI or Hoechst for nuclear visualization
Consider double-staining with mitochondrial markers when studying mitochondrial functions
Capture z-stacks to accurately assess nuclear versus cytoplasmic distribution
Include single-stained controls for spectral bleed-through correction
Compare localization before and after cellular stressors (e.g., serum deprivation or staurosporine treatment)
Document time-dependent changes in localization following stress induction
ATF5 antibodies have become valuable tools in cancer research due to ATF5's role in cancer cell survival and chemoresistance:
IHC analysis has revealed that ATF5 is highly expressed in glioblastomas, breast cancers, and pancreatic carcinomas compared to normal tissues
Quantitative analysis correlating ATF5 expression with patient outcomes can help establish prognostic value
Studies in pancreatic cancer cells demonstrated that ATF5 interference enhanced sensitivity to paclitaxel by modulating BCL-2 and BAX expression
Western blot analysis with ATF5 antibodies can quantify changes in expression following drug treatments
ATF5 antibodies can confirm knockdown efficiency in experiments using siRNA or shRNA approaches
In a study on mammary tumors, ATF5 knockdown resulted in decreased tumor volume and weight, reduced proliferation rate, and diminished migratory potential
ChIP assays using ATF5 antibodies have identified direct binding to the ARE region of the Egr-1 promoter, revealing a molecular mechanism for ATF5's role in cell proliferation
Co-immunoprecipitation with ATF5 antibodies has uncovered interactions with p300, revealing regulatory mechanisms involving acetylation
ATF5 undergoes several post-translational modifications that regulate its function:
Immunoprecipitate ATF5 with a specific antibody, followed by Western blotting with anti-acetyl-lysine antibodies
Alternatively, use acetylation site-specific antibodies when available
Treatment with deacetylase inhibitors like trichostatin A (TSA) enhances detection of acetylated ATF5
Use phospho-specific antibodies when available
Alternatively, perform immunoprecipitation with ATF5 antibodies followed by Western blotting with anti-phospho-serine/threonine/tyrosine antibodies
Lambda phosphatase treatment can serve as a control to validate phosphorylation signals
Immunoprecipitate ATF5 under denaturing conditions (to disrupt protein-protein interactions)
Western blot with anti-ubiquitin antibodies
Use proteasome inhibitors (MG132) to enhance detection of ubiquitinated forms
Immunoprecipitate ATF5 and subject to tryptic digestion
LC-MS/MS analysis can identify multiple modifications simultaneously
SILAC approaches can quantify changes in modification states under different conditions
ATF5 plays a crucial role in mitochondrial quality control, particularly in response to stress:
Use ATF5 antibodies to track ATF5 translocation to mitochondria during stress
Co-staining with mitochondrial markers can confirm localization
RNA-seq analysis in ATF5 knockdown cells has revealed that ATF5 regulates genes involved in the UPRmt pathway
Studies have shown that ATF5 knockdown decreases mitochondrial membrane potential and ATP levels
Measure these parameters using fluorescent dyes (JC-1, TMRE) for membrane potential and luciferase-based assays for ATP
ATF5 knockdown impacts mitochondrial fusion and fission by regulating genes like Drp1, Fis-1, Opa1, Mfn1, and Mfn2
Use immunofluorescence with ATF5 antibodies alongside mitochondrial morphology assessments
ATF5 knockdown increases autophagy as measured by LC3B immunofluorescence
Use ATF5 antibodies in combination with autophagy markers to assess the relationship between ATF5 and mitochondrial clearance
Multiple bands in ATF5 Western blots can occur for several legitimate reasons:
Acetylated ATF5 (particularly at K29) may show altered migration
Phosphorylated forms can appear as higher molecular weight bands
Ubiquitinated ATF5 will appear as a ladder of higher molecular weight bands
Different ATF5 isoforms may be expressed in certain tissues or under specific conditions
Verify these are legitimate isoforms by comparing with literature reports
C-terminal fragments may be detected if the antibody recognizes this region
Improve sample preparation by adding protease inhibitors and keeping samples cold
Some antibodies may detect related ATF family members (particularly ATF4, an important paralog)
Use ATF5 knockout or knockdown samples as negative controls to confirm specificity
Run a gradient gel to better separate closely migrating bands
Perform peptide competition assays to identify specific versus non-specific bands
Pre-adsorb antibodies with recombinant ATF5 protein to reduce non-specific binding
Rigorous controls are critical for accurate interpretation of ATF5 functional studies:
Include multiple siRNA/shRNA sequences targeting different regions of ATF5 to rule out off-target effects
Confirm knockdown efficiency at both mRNA (qRT-PCR) and protein levels (Western blot)
Include a non-targeting siRNA/shRNA control with similar chemical modifications
In the study by Frontiers in Endocrinology, researchers systematically validated their knockdown approach before investigating tumor volume effects
Include empty vector controls processed identically to ATF5-expressing samples
Consider using an inducible expression system to control expression levels
Tag location (N- vs. C-terminal) may affect function; validate function using known ATF5 activities
Express siRNA/shRNA-resistant ATF5 to demonstrate specificity of observed phenotypes
Include non-functional ATF5 mutants (e.g., K29R mutant that cannot be acetylated) as negative controls
Assess known ATF5 transcriptional targets like Egr-1 using reporter assays
Measure changes in mitochondrial parameters known to be affected by ATF5 modulation
Document changes in downstream protein expression (e.g., BCL-2, BAX)
ATF5 exhibits context-dependent functions that may appear contradictory:
ATF5 is essential for survival in cancer cells but dispensable in non-transformed cells
In olfactory sensory neurons, ATF5 is required for terminal differentiation and survival
Document the baseline expression levels of ATF5 and its binding partners in your model system
ATF5 response may differ dramatically between basal and stress conditions
The physical association between ATF5 and p300 is disrupted after serum deprivation or staurosporine treatment
Design experiments that explicitly compare multiple stress conditions with appropriate controls
Absence of ATF5 triggers compensatory increases in PGC-1α and ATF4 expression
Consider analyzing related family members and pathway components when manipulating ATF5
Create a systematic comparison table of experimental conditions across studies
Perform time-course experiments to capture dynamic responses
Use genetic approaches (CRISPR/Cas9) for complete knockout versus transient knockdown to distinguish acute versus chronic effects
Consider multiple functional outputs simultaneously (e.g., proliferation, survival, mitochondrial function)
Use systems biology approaches to model ATF5 in its network context
When possible, validate in vitro findings with in vivo models to assess physiological relevance
Recent research has established ATF5 as a regulator of exercise-induced mitochondrial quality control:
Studies have shown that lack of ATF5 increases mitochondrial content but reduces function
Use ATF5 antibodies to correlate protein levels with mitochondrial parameters measured by complementary techniques
ATF5 is required for normal mitophagic and UPRmt mRNA responses to exercise
Design protocols that compare sedentary versus exercise conditions with appropriate timing of sample collection
Combine ATF5 immunoprecipitation with mass spectrometry to identify exercise-induced binding partners
Develop ChIP-seq protocols using ATF5 antibodies to identify genomic binding sites that respond to exercise
Employ proximity ligation assays to visualize interactions between ATF5 and mitochondrial proteins
Investigate whether exercise-induced improvements in mitochondrial health are ATF5-dependent
Explore small molecule modulators of ATF5 and test effects on mitochondrial quality control