Gene ID: HGNC: 28915; NCBI Gene: 55509; Ensembl: ENSG00000123685; UniProt: Q9NR55 .
Protein Structure:
BATF3 is indispensable for the differentiation of CD8α⁺ conventional dendritic cells (cDCs) and CD103⁺ dendritic cells, which are critical for cross-presenting antigens to CD8⁺ T-cells and producing IL-12 during infections .
Th9 Differentiation: BATF3 enhances IL-9 production in Th9 cells by forming a complex with IRF4, promoting allergic inflammation .
Treg Suppression: BATF3 inhibits regulatory T-cell (Treg) differentiation by binding to the Foxp3 locus (CNS1 region), skewing immune responses toward effector T-cells .
Memory CD8⁺ T-Cells: BATF3 promotes survival of CD8⁺ T-cells by suppressing BIM-mediated apoptosis, enhancing long-term memory .
Th9 Differentiation (Nature, 2019):
Treg Inhibition (Nature, 2017):
Metabolic Syndrome (Frontiers, 2022):
Cancer Immunotherapy: BATF3-driven cDC1s enhance antitumor immunity by priming CD8⁺ T-cells .
Autoimmune Diseases: Targeting BATF3 could modulate Treg/Teffector balance in conditions like IBD .
Infection Control: BATF3 deficiency impairs containment of pathogens (e.g., Cryptococcus neoformans) in lungs and brain .
BATF3 (Basic Leucine Zipper ATF-Like Transcription Factor 3) is a 14.5-20 kDa nuclear member of the bZIP family of proteins. It functions as a transcription factor that controls the differentiation of CD8+ conventional dendritic cells (cDCs) in the immune system. BATF3 acts via formation of heterodimers with JUN family proteins, recognizing and binding DNA sequence 5'-TGA[CG]TCA-3' to regulate target gene expression. In humans, BATF3 is preferentially expressed in effector CD4 T cells and conventional dendritic cells, where it plays critical roles in immune response regulation and cell fate determination .
BATF3 shows differential expression across immune cell populations. It is predominantly expressed in:
Effector CD4 T cells but not in regulatory T (Treg) cells
CD141+ Myeloid Dendritic Cells
CD8α+ Thymic Conventional Dendritic Cells
Conventional dendritic cells (CD11c+)
Th1 cells
The expression is typically transient in T cells, appearing within the first days after T cell priming and having long-lasting effects. Detection methods such as flow cytometry have shown BATF3 expression in human PBMC monocytes, particularly in CD11c+ populations, and Jurkat T cell leukemia cell lines .
BATF3 is essential for the development of conventional type 1 dendritic cells (cDC1), specifically controlling the differentiation of CD8α+ and CD103+ DCs. These BATF3-dependent cDC1s are critical for priming CD8+ T cell-mediated immunity against intracellular pathogens and malignancies.
To study this function, researchers can:
Use Batf3-knockout mouse models that specifically lack cDC1 populations
Perform lineage-tracing experiments using cell-specific markers (CD103, CD8α, XCR1)
Implement conditional knockout systems to temporally control BATF3 deletion
Conduct bone marrow chimera experiments to assess cell-intrinsic effects
Analyze human BATF3 polymorphisms and correlate with dendritic cell phenotypes .
BATF3 programs CD8+ T cell memory through multiple mechanisms:
When overexpressed in CD8+ T cells, BATF3 enhances their survival and transition to memory state
BATF3 regulates T cell apoptosis and longevity via the proapoptotic factor BIM
BATF3-expressing T cells show normal expansion and differentiation but reduced contraction phase
Gene Ontology analyses revealed that BATF3-induced genes are enriched for DNA and mRNA metabolic processing, ribosomal biogenesis and metabolic pathways such as glycolysis
BATF3 overexpression increases expression of IL7R, a surface marker associated with T cell survival, long-term persistence, and positive clinical response to adoptive cell therapy. Mechanistically, BATF3 overexpression increases chromatin accessibility at regions near memory-associated genes (TCF7, MYB, IL7R, CCR7, SELL) while reducing accessibility at exhaustion-associated loci (TIGIT, CTLA4, LAG3) .
BATF3 acts as a fate-decision factor in T cell differentiation by inhibiting regulatory T cell development. Key findings include:
BATF3 is preferentially expressed in effector CD4 T cells but not in Treg cells
Ectopic expression of BATF3 inhibits Foxp3 induction, the master transcription factor for Treg differentiation
BATF3-deficient CD4 T cells preferentially differentiate into Treg cells in vitro and in vivo
Mechanistically, BATF3 binds to the CNS1 region of the Foxp3 locus and reduces its expression
BATF3 knockout mice show enhanced Treg function in gut-associated immune disease models
This inhibitory role positions BATF3 as a critical regulator of the balance between effector and regulatory T cell populations .
For detecting and quantifying BATF3 in human samples, researchers can use several complementary approaches:
Method | Applications | Considerations | Detection Sensitivity |
---|---|---|---|
Western Blot | Protein level detection | BATF3 detected at ~17 kDa; use reducing conditions | Moderate |
Flow Cytometry | Single-cell analysis | Requires cell fixation/permeabilization; best with anti-BATF3 monoclonal antibodies | High |
Immunohistochemistry | Tissue localization | Paraffin-embedded sections work well; nuclear localization visible | Moderate |
RT-qPCR | mRNA quantification | Design primers spanning exon junctions; normalize to stable reference genes | High |
ChIP-seq | DNA binding analysis | Identify BATF3 binding sites; requires high-quality antibodies | Moderate-High |
RNA-seq | Transcriptome analysis | Reveals BATF3-regulated genes | Very High |
For optimal results, intracellular staining protocols with proper fixation and permeabilization are essential when using flow cytometry. Both polyclonal and monoclonal antibodies are available, with monoclonals offering better specificity for human BATF3 .
Several approaches are available for manipulating BATF3 expression in primary human T cells:
Lentiviral/Retroviral Expression Systems:
Compact size of BATF3 (381 bp) makes it amenable to viral delivery
Can be incorporated into the same lentivirus delivering CAR or TCR constructs
Consider including tracking markers (GFP) to identify transduced cells
CRISPR-Based Methods:
CRISPRa for gene activation: dSaCas9-based systems can upregulate endogenous BATF3
CRISPRi for gene repression: Target BATF3 promoter regions
CRISPRko for complete knockout: Multiple guide RNAs targeting different exons
Transfection Approaches:
Electroporation of BATF3 mRNA for transient expression
Nucleofection of plasmid DNA for longer expression
Inducible Systems:
Tet-On/Off systems for temporal control of BATF3 expression
Destabilization domain-based systems for rapid protein depletion
For optimal results, researchers should consider timing of expression manipulation relative to T cell activation state, as BATF3 effects are context-dependent .
When using BATF3 knockout models, researchers should consider:
Developmental vs. Acute Effects:
Constitutional knockouts affect development of cDC1s, causing broad immunological defects
Consider conditional/inducible knockouts to separate developmental from functional effects
Cell Type-Specific Consequences:
Primary defect is in CD8α+ and CD103+ dendritic cell development
Secondary effects on CD8+ T cell priming and memory formation
Altered IgA-coating of bacteria and microbial dysbiosis
Compensatory Mechanisms:
Alternative transcription factors may partially compensate for BATF3 deficiency
Assess expression of related factors (BATF, BATF2, AP-1 family members)
Strain and Environmental Considerations:
Different mouse strains may show variable phenotype severity
Microbiome composition significantly impacts phenotype (consider co-housing controls)
Diet interactions are particularly important (high-fat diet enhances metabolic phenotypes)
Experimental Design Controls:
BATF3 contributes to anti-tumor immunity through multiple mechanisms:
Development of Critical Dendritic Cell Populations:
BATF3 is essential for the development of cDC1s (CD103+ DCs) that cross-present tumor antigens to CD8+ T cells
BATF3-dependent DCs are required within the tumor microenvironment (TME) for PD-1/PD-L1 blockade efficacy
T Cell Programming Effects:
BATF3 overexpression in CD8+ T cells enhances their survival and transition to memory state
Decreases expression of exhaustion-associated genes like TIGIT, CTLA4, and LAG3
Programs a transcriptional profile that correlates with positive clinical response to adoptive T cell therapy
Costimulatory Functions:
BATF3+ DCs provide critical 4-1BB/4-1BBL costimulatory signals within the tumor microenvironment
These signals are required for the efficacy of checkpoint blockade therapy
For therapeutic applications, BATF3 overexpression markedly enhanced the potency of CAR T cells in both in vitro and in vivo tumor models. Its compact size makes it particularly amenable to integration into current adoptive cell therapy manufacturing processes .
BATF3 plays a protective role against metabolic syndrome and maintains intestinal epithelial homeostasis:
Metabolic Regulation:
BATF3-deficient mice develop metabolic syndrome characterized by insulin resistance, elevated blood glucose and serum insulin levels, increased body weight and white adipocyte size
Hyperinsulinemia and hypercholesterolemia are the earliest metabolic changes observed in lean BATF3-deficient mice
Intestinal Barrier Function:
BATF3 deficiency leads to altered localization of tight junction proteins (occludin-1, ZO-1, claudin-2) in intestinal epithelial cells
This results in increased intestinal permeability, which contributes to low-grade inflammation
Treatment with glycolysis inhibitor 2-deoxy-D-glucose reduced intestinal inflammation and restored barrier function
Microbiome Effects:
BATF3-deficient mice show decreased IgA-coating of fecal bacteria
Display microbial dysbiosis with decreased levels of beneficial bacteria like Akkermansia muciniphila and Bifidobacterium
Antibiotic treatment prevents metabolic syndrome development in these mice
These findings suggest that BATF3-dependent dendritic cells play a critical role in maintaining intestinal barrier function and preventing metabolic dysregulation through microbiome-dependent mechanisms .
BATF3 functions through a complex network of interactions with other transcription factors:
JUN Family Interactions:
BATF3 heterodimerizes with JUN family proteins to bind DNA
Acts as a transcriptional repressor when heterodimerizing with JUN
May function in repression of interleukin-2 and matrix metalloproteinase-1 transcription
IRF4 Cooperative Activity:
Forms a BATF3/IRF4 complex that can bind the IL-9 promoter to induce Th9 cell differentiation
CRISPRko screens identified IRF4 as a cofactor of BATF3 in T cells
Glucocorticoid Receptor Interactions:
BATF3 acts as a gene-specific coactivator of the Glucocorticoid Receptor (GR)
This coactivator potency is influenced by the sequence of the GR binding site
Interaction with GR is modulated by the GR "lever arm" domain
JUNB Cooperation:
CRISPRko screens revealed that BATF3 heterodimerizes with JUNB in CD8+ T cells
This interaction is essential for driving transcriptional programs
Understanding these interactions is crucial for developing strategies to modulate BATF3 activity in therapeutic contexts .
BATF3 exerts significant effects on chromatin accessibility and epigenetic regulation:
Chromatin Accessibility Changes:
BATF3 overexpression increases accessibility at regions near memory-associated genes (TCF7, MYB, IL7R, CCR7, SELL)
Decreases accessibility at exhaustion-associated loci (TIGIT, CTLA4, LAG3)
Approximately 25% of genes that change expression with BATF3 overexpression show corresponding changes in local chromatin accessibility
DNA Binding Patterns:
BATF3 binds to the CNS1 region of the Foxp3 locus to reduce its expression
Acts by recognizing and binding the DNA sequence 5'-TGA[CG]TCA-3'
Methodological Approaches:
Technique | Application | Key Considerations |
---|---|---|
ATAC-seq | Maps genome-wide chromatin accessibility | 50,000 cells minimum; sort for viable cells |
ChIP-seq | Identifies direct BATF3 binding sites | Requires high-quality ChIP-grade antibodies |
CUT&RUN | Alternative to ChIP with lower cell numbers | Better signal-to-noise ratio than ChIP |
HiChIP | Identifies long-range chromatin interactions | Maps 3D chromatin organization at BATF3 loci |
CRISPRi-seq | Screens for functional impact of BATF3 binding | Requires dCas9-KRAB targeting to BATF3 sites |
HOMER motif analysis can be used to find transcription factor binding motifs that contribute to changes in chromatin accessibility with BATF3 overexpression compared to control cells .
Current challenges and future directions in BATF3 research include:
Safety Considerations for Therapeutic Applications:
Sustained expression of BATF3 combined with other genetic alterations could potentially lead to antigen-independent clonal T cell expansion
Need to develop transient delivery methods, modulate transgene expression, or integrate suicide switches
Cell Type-Specific Functions:
Better understanding of BATF3 roles beyond dendritic cells and T cells
Context-dependent functions in different tissue microenvironments
Temporal Dynamics:
Elucidating the temporal regulation of BATF3 expression during immune responses
Understanding how transient expression yields long-lasting effects
Tissue-Specific Effects:
Exploring BATF3 function in specialized tissue niches (e.g., tumor microenvironment, intestinal mucosa)
Differences between circulating and tissue-resident immune cells
Systems Biology Approaches:
Integration of multi-omics data to build comprehensive models of BATF3 regulatory networks
Single-cell approaches to address cellular heterogeneity
Therapeutic Engineering:
Developing optimized BATF3 variants for enhanced T cell persistence in adoptive therapies
Combining BATF3 with other transcription factors for synergistic effects
Metabolic Integration:
The Basic Leucine Zipper Transcription Factor ATF-Like 3 (BATF3) is a member of the basic leucine zipper (bZIP) protein family. This family of proteins is characterized by the presence of a leucine zipper motif that facilitates dimerization and a basic region that binds to DNA. BATF3 is a transcription factor that plays a crucial role in the regulation of gene expression, particularly in the immune system.
The BATF3 gene is located on chromosome 1 in humans and encodes a protein that is approximately 21 kDa in size . The protein contains a basic leucine zipper domain, which is essential for its function as a transcription factor. This domain allows BATF3 to form heterodimers with other proteins, such as members of the JUN family, to regulate the transcription of target genes .
BATF3 is primarily known for its role in the immune system. It is a key regulator of the differentiation of CD8+ dendritic cells (DCs), which are essential for the immune response against pathogens . These dendritic cells are involved in the presentation of antigens to CD8+ T cells, leading to the activation of these T cells and the production of interleukin-12 (IL-12), a cytokine that is crucial for the immune response .
BATF3 functions by forming heterodimers with JUN family proteins. These heterodimers bind to specific DNA sequences, known as AP-1 sites, to regulate the transcription of target genes . The BATF3-JUN heterodimer can also interact with other transcription factors, such as IRF4 or IRF8, to form a complex that binds to immune-specific regulatory elements and activates the expression of genes involved in the immune response .