TAF15 belongs to the FET protein family (FUS, EWSR1, TAF15) and shares conserved domains:
N-terminal QGSY-rich domain: Facilitates transcriptional activation and protein-protein interactions .
RNA Recognition Motif (RRM): Mediates sequence-specific RNA binding, preferentially targeting GGUA motifs .
C-terminal Zinc Finger (ZnF) and RGG motifs: Enable nucleic acid binding and phase separation .
Structural studies reveal that TAF15 forms amyloid filaments in neurodegenerative diseases, with residues 7–99 adopting a β-sheet-rich fold stabilized by hydrogen-bonding networks .
As a subunit of TFIID, TAF15 anchors RNA polymerase II to promoter regions, enabling transcription initiation .
Interacts with POLR2C, POLR2E, and TATA-binding protein (TBP) to stabilize the pre-initiation complex .
Binds ~4,900 RNAs in mouse brains, primarily in introns and 3′ UTRs, influencing mRNA stability and transport .
Associates with U1 snRNA to form a chromatin-linked snRNP, potentially regulating spliceosome activity .
TAF15 is overexpressed in multiple cancers and drives tumor progression via the RAF1/MEK/ERK pathway:
TAF15, FUS, and TDP-43 exhibit overlapping RNA targets but distinct splicing roles. TAF15 depletion alters mRNA turnover in motor neurons, mimicking ALS-associated FUS mutations .
TAF15 forms amyloid filaments with a β-strand-rich fold (residues 7–99) in FTLD patients. These aggregates are structurally distinct from tau or TDP-43 pathologies .
In Xenopus, TAF15 is essential for dorsoanterior neural tissue development, regulating genes like otx2 and pax6 .
Maternally deposited and upregulated post-zygotic genome activation .
Biomarker Potential: Overexpression in GIST and GC correlates with advanced stages .
Therapeutic Targeting: siRNA-mediated TAF15 knockdown reduces tumor growth by 60% in xenograft models .
Neurodegeneration: Cryo-EM structures of TAF15 filaments enable targeted drug screening for FTLD .
Mechanistic links between TAF15’s transcriptional and RNA metabolic roles.
Role of TAF15 amyloid filaments in cross-seeding other proteinopathies.
Tissue-specific isoforms and post-translational modifications.
TAF15 (TATA-box binding protein-associated factor 2N) is an RNA-binding protein belonging to the FET family, which also includes FUS (Fused in Sarcoma) and EWS (Ewing Sarcoma). In normal cellular physiology, TAF15 participates in transcriptional regulation and RNA processing. Studies have shown that TAF15 binds to approximately 4,900 RNA targets enriched for GGUA motifs in adult mouse brains .
Methodologically, researchers characterize TAF15's normal function through:
RNA binding assays (CLIP-seq and RNA Bind-N-Seq technologies)
Protein interaction studies using immunoprecipitation
Transcriptomic analyses following TAF15 knockdown or knockout
Unlike its related protein FUS, TAF15 appears to have a minimal role in alternative splicing but contributes significantly to RNA turnover in human neural progenitors .
TAF15 shares structural similarities with other FET proteins (FUS and EWS), particularly in its domain organization:
| Domain | TAF15 | FUS | EWS | Function |
|---|---|---|---|---|
| Low-complexity domain (LCD) | Present (N-terminal) | Present (N-terminal) | Present (N-terminal) | Phase separation; amyloid formation |
| RNA recognition motif (RRM) | Present | Present | Present | RNA binding |
| Zinc finger domain | Present | Present | Present | Nucleic acid interaction |
| Nuclear localization signal (NLS) | Present | Present | Present | Nuclear import |
The filamentous structure of TAF15 in disease states involves residues 7-99 in the low-complexity domain, forming a characteristic 13-β-strand fold . This structure is distinct from filaments formed by other amyloidogenic proteins like tau or TDP-43, suggesting unique pathological mechanisms .
Researchers typically study these structural features through:
Cryo-electron microscopy (cryo-EM) for filament structures
X-ray crystallography for domain structure determination
NMR spectroscopy for dynamic structural elements
Recent breakthrough research has identified TAF15 as the key protein forming abnormal aggregates in a subset of frontotemporal dementia cases. Specifically:
Cryo-EM studies revealed abundant TAF15 amyloid filaments in the brains of individuals with frontotemporal lobar degeneration (FTLD), particularly in cases previously designated as FTLD-FUS .
The filament structures were consistent across multiple patients and brain regions, suggesting a specific pathological process .
TAF15 filaments were found in the absence of FUS filaments, despite FUS often being present in the same inclusions .
In approximately 10% of frontotemporal dementia cases where the causative protein was previously unknown, TAF15 has now been identified as the primary amyloidogenic protein .
Methodologically, researchers established this link through:
Post-mortem brain tissue analysis using immunohistochemistry
Protein extraction and characterization from affected brain regions
Cryo-EM structural analysis of protein filaments
Mass spectrometry verification of protein identity
TAF15 pathology shows both similarities and differences in frontotemporal dementia and motor neuron diseases:
| Feature | Frontotemporal Dementia | Motor Neuron Disease |
|---|---|---|
| TAF15 filament structure | 13-β-strand fold from residues 7-99 | Identical fold when present |
| Brain regions affected | Prefrontal and temporal cortices | Motor cortex and brainstem |
| Co-pathology | May include TMEM106B and Aβ filaments | Less frequently observed |
| Clinical correlation | Primary cognitive/behavioral symptoms | Motor deficits, may precede FTD symptoms |
In studies of individuals with both upper and lower motor neuron pathology, TAF15 filaments with identical structure to those found in prefrontal and temporal cortices were observed in motor regions . This suggests that TAF15 proteinopathy may exist on a disease spectrum encompassing both frontotemporal dementia and motor neuron disease, analogous to the TDP-43 pathology spectrum .
Research approaches include:
Comparative neuropathological analyses across disease phenotypes
Regional protein extraction and filament characterization
Clinicopathological correlation studies
Animal modeling of region-specific TAF15 pathology
Detecting and characterizing TAF15 aggregates requires a multi-modal approach:
Immunohistochemistry/Immunofluorescence:
Primary antibodies: Anti-TAF15 (specifically recognizing epitopes outside the filament core)
Co-staining with FUS and transportin 1 antibodies to characterize inclusion composition
Controls should include known TAF15-positive cases and neurologically normal individuals
Biochemical Extraction and Analysis:
Sequential extraction using increasing detergent strengths
Sarkosyl-insoluble fraction isolation for filamentous proteins
Western blotting with TAF15-specific antibodies
Mass spectrometry confirmation focusing on peptides mapping to the filament core region (residues 7-99)
Structural Analysis:
Quantification Methods:
Stereological counting of TAF15-positive inclusions
Digital image analysis of immunostained sections
Regional distribution mapping correlating with clinical phenotypes
Developing accurate models for TAF15 pathology remains challenging. Current approaches include:
| Model Type | Advantages | Limitations | Key Considerations |
|---|---|---|---|
| Primary neuron cultures | Human cellular context; accessible | Limited lifespan; lacks systemic factors | TAF15 overexpression or knockdown possible |
| iPSC-derived neurons | Patient-specific; developmental processes | Variability; maturation challenges | Can model both TAF15 loss and aggregation |
| Drosophila | Rapid generation; behavioral assessment | Evolutionary distance | Focus on conserved domains |
| Mouse models | Mammalian brain; behavioral testing | Species differences in TAF15 biology | Requires careful design of transgenic approach |
For cellular models, researchers should consider:
Using human iPSC-derived motor neurons or cortical neurons
Creating isogenic lines with TAF15 mutations or altered expression
Evaluating both TAF15 loss-of-function and aggregation phenotypes
Assessing RNA metabolism changes through transcriptomic approaches
For animal models, key methodological considerations include:
Targeting expression to relevant neuronal populations
Using brain region-specific inducible systems
Validating models against human neuropathological findings
Assessing both biochemical and behavioral phenotypes
TAF15, FUS, and TDP-43 show distinct RNA binding patterns and functional consequences:
| Feature | TAF15 | FUS | TDP-43 |
|---|---|---|---|
| RNA motif preference | GGUA-rich sequences | GGU-rich sequences | GU-rich sequences |
| Binding location | Introns and 3' UTRs | Introns and 3' UTRs | Introns and 3' UTRs |
| Effect on splicing | Minimal role | Significant regulator | Significant regulator |
| Effect on RNA turnover | Regulates target stability | Regulates target stability | Regulates stability and processing |
| Number of RNA targets | ~4,900 in mouse brain | ~5,500 in mouse brain | ~6,000 in mouse brain |
Research approaches to characterize these differences include:
CLIP-seq (Cross-linking immunoprecipitation followed by sequencing)
RNA Bind-N-Seq for motif identification
RNA stability assays following protein knockdown
Alternative splicing analysis using RNA-seq
Direct comparison studies in the same cellular systems
These differences suggest both overlapping and distinct pathological mechanisms when each protein forms aggregates in disease states .
Studying TAF15-RNA interactions in disease contexts requires specialized approaches:
Tissue-based RNA-protein interaction studies:
CLIP-seq adapted for post-mortem human brain tissue
Isolation of pathological inclusions followed by RNA extraction and sequencing
RNAscope in situ hybridization combined with TAF15 immunostaining
Single-cell approaches to capture cell-type specific interactions
Biochemical approaches:
RNA immunoprecipitation from disease models
In vitro RNA binding assays with recombinant normal and mutant TAF15
Competition assays between TAF15, FUS, and other RNA-binding proteins
Structural studies of TAF15-RNA complexes
Functional assessments:
RNA stability assays following TAF15 aggregation
Alternative splicing analysis in disease models
Translation efficiency measurements
RNA localization studies in the presence of TAF15 aggregates
Advanced techniques:
APEX-seq for proximity labeling of RNAs near TAF15 aggregates
Single-molecule imaging of TAF15-RNA interactions
Massively parallel reporter assays for TAF15 binding sites
RNA modifications analysis in relation to TAF15 binding
The high-resolution structure of TAF15 filaments (2.0-2.7 Å) provides crucial insights for therapeutic development:
Structure-based drug design opportunities:
The 13-β-strand fold comprising residues 7-99 of TAF15 presents specific targets for small molecule binding
Stabilizing compounds that prevent further aggregation
Destabilizing agents that could promote disaggregation
Compounds targeting specific structural motifs unique to TAF15 filaments
Immunotherapeutic approaches:
Generation of conformation-specific antibodies recognizing TAF15 filaments
Screening epitopes outside the filament core for accessible antibody targets
Developing intrabodies targeting pre-fibrillar species
Diagnostic applications:
PET ligands based on TAF15 filament binding properties
CSF biomarkers detecting TAF15 fragments or conformations
Blood-based assays for TAF15 pathology
Methodological considerations:
In silico screening against specific pockets in the TAF15 filament structure
Fragment-based drug discovery targeting the filament interface
Validation in cellular models expressing the same filament structure
Pharmacokinetic optimization for CNS penetration
This approach mirrors strategies already being developed for tau and Aβ in Alzheimer's disease , but requires TAF15-specific optimization.
TAF15 aggregation and nuclear-cytoplasmic transport disruption appear interconnected:
Evidence for transport disruption:
Potential mechanisms:
Sequestration of transportin 1 by TAF15 aggregates via NLS interactions
Competition between normal and aggregated TAF15 for transport machinery
Disruption of nuclear pore complex function by TAF15 aggregates
Secondary effects on nucleocytoplasmic transport of other essential cargoes
Experimental approaches:
Live cell imaging of nuclear transport in TAF15 pathology models
Selective inhibition of transportin-mediated import
Rescue experiments restoring nuclear localization
Nucleocytoplasmic fractionation studies in disease models
Therapeutic implications:
Targeting the TAF15-transportin 1 interaction
Enhancing nuclear import efficiency
Preventing cytoplasmic aggregation through chaperone enhancement
Combinatorial approaches addressing both aggregation and transport defects
This represents a critical area for future research, as nuclear-cytoplasmic transport defects may constitute both a consequence and driver of disease progression.
TAF15 joins a select group of proteins forming amyloid filaments in neurodegenerative diseases:
| Protein | Diseases | Filament Structure | Cell Types Affected | Unique Features |
|---|---|---|---|---|
| TAF15 | FTLD-FET | 13-β-strand fold from LCD | Neurons in frontal/temporal cortex; motor neurons | Co-localization with FUS |
| FUS | ALS-FUS | Not definitively identified in human brain | Motor neurons; frontal cortex neurons | RNA processing defects |
| TDP-43 | ALS-TDP; FTLD-TDP | Identified in ALS | Motor neurons; frontal cortex neurons | C-terminal fragments common |
| Tau | AD; FTLD-tau; PSP; CBD | Multiple distinct folds | Neurons; some glial involvement | Strain-specific pathology |
| Aβ | AD | Multiple polymorphs | Extracellular deposition | Plaque formation |
| α-synuclein | PD; DLB; MSA | Multiple polymorphs | Neurons; oligodendrocytes (MSA) | Lewy body formation |
TAF15 proteinopathy shows distinct characteristics:
Forms amyloid filaments specifically in FTLD-FET cases
Involves an RNA-binding protein with specific RNA targets
Co-occurs with other FET proteins in inclusions without them forming filaments
Appears to have a disease spectrum spanning cognitive and motor phenotypes
Methodologically, researchers compare these proteinopathies through:
Cryo-EM structural studies of isolated filaments
Biochemical characterization of aggregation propensities
Cellular models of seeding and spreading
Comparative neuropathological analyses
Despite recent advances, significant questions remain about FET protein interactions in disease:
Sequestration dynamics:
Why do TAF15 filaments form while FUS is present in inclusions but does not form filaments?
What molecular interactions determine which FET protein forms filaments?
Is there a protective mechanism preventing FUS fibrillization despite its aggregation propensity?
Cross-seeding potential:
Can TAF15 filaments seed FUS or EWS aggregation?
Do subclinical aggregates of multiple FET proteins exist?
What determines the dominant protein in different disease contexts?
Functional consequences:
How does loss of multiple FET proteins compare to single protein dysfunction?
Are there compensatory mechanisms between FET proteins?
Do FET proteins show cell-type specific vulnerabilities?
Methodological approaches needed:
Development of FET protein-specific antibodies distinguishing native and pathological conformations
Cellular models expressing physiological levels of all FET proteins
Advanced imaging techniques to observe early aggregation events
Cross-species comparisons of FET protein interactions
The recent identification of TAF15 filaments challenges previous assumptions about FTLD-FUS and necessitates reevaluation of the role of each FET protein in disease .
Several genetic factors may influence TAF15 aggregation:
Direct genetic factors:
Variations in the TAF15 gene, particularly in the low-complexity domain (residues 7-99)
Mutations affecting TAF15 post-translational modifications
Alterations in nuclear localization signal or RNA-binding domains
Genetic modifiers of TAF15 expression levels
Indirect genetic influences:
Genes involved in protein quality control (chaperones, degradation pathways)
RNA processing machinery components
Nuclear transport factors (transportin 1, nucleoporins)
Stress response mediators affecting phase separation properties
Research approaches:
Genome-wide association studies in FTLD cohorts
Whole exome/genome sequencing of FTLD-FET cases
CRISPR screens for modifiers of TAF15 aggregation
Targeted sequencing of candidate genetic modifiers
Translational potential:
Identification of at-risk individuals through genetic screening
Discovery of novel therapeutic targets
Development of personalized intervention strategies
Understanding of disease heterogeneity
A systematic evaluation of these factors would significantly advance our understanding of why TAF15 forms pathological aggregates in specific individuals.
Development of TAF15-targeted therapies requires systematic approaches:
Therapeutic strategies:
Antisense oligonucleotides (ASOs) reducing TAF15 expression
Small molecules preventing TAF15 aggregation or promoting clearance
Gene therapy approaches to modulate TAF15 levels or function
Immunotherapies targeting pathological TAF15 conformations
Preclinical model development:
iPSC-derived neurons expressing TAF15 with enhanced aggregation propensity
Transgenic animals expressing human TAF15 in vulnerable neuronal populations
Viral vector-mediated TAF15 pathology induction
Ex vivo models using organotypic brain slices
Evaluation methodologies:
Biochemical assessment of TAF15 aggregation and solubility
Imaging of TAF15 localization and inclusion formation
Transcriptomic analysis of TAF15-regulated RNA targets
Functional assessment of neuronal health and activity
Behavioral testing in animal models
Translational considerations:
Development of target engagement biomarkers
Establishment of pharmacodynamic readouts
Determination of therapeutic window
Assessment of safety profile considering TAF15's normal functions
These approaches require close collaboration between structural biologists, medicinal chemists, neuroscientists, and clinicians to advance potential TAF15-directed therapies toward clinical application.
Single-cell technologies offer unique opportunities to understand TAF15 pathology:
Single-cell transcriptomics:
Identification of cell-type specific responses to TAF15 aggregation
Characterization of transcriptional changes in individual affected neurons
Detection of compensatory mechanisms in resistant cells
Mapping disease progression at cellular resolution
Single-cell proteomics:
Quantification of TAF15 protein levels in individual cells
Identification of protein interaction networks in affected vs. unaffected cells
Detection of post-translational modifications influencing aggregation
Assessment of protein quality control responses
Spatial transcriptomics/proteomics:
Mapping TAF15 pathology in specific brain regions and circuits
Correlation with regional vulnerability patterns
Identification of microenvironmental factors influencing pathology
Assessment of non-cell autonomous effects
Methodological considerations:
Sample preparation preserving cellular integrity and protein state
Analysis pipelines integrating multiple single-cell modalities
Validation in human post-mortem tissue and disease models
Computational approaches for trajectory analysis of disease progression
These technologies can reveal heterogeneity in TAF15 pathology that may be obscured in bulk tissue analyses, potentially identifying new therapeutic targets and biomarkers.
Development of TAF15 pathology biomarkers represents a critical research need:
Cerebrospinal fluid (CSF) biomarkers:
Total TAF15 protein levels in CSF
TAF15 fragments or modified forms specific to pathological states
Conformation-specific assays detecting misfolded TAF15
Ratios of TAF15 to other FET proteins
Neuroimaging approaches:
PET ligands targeting TAF15 filaments
Functional MRI correlates of TAF15-associated network dysfunction
Structural imaging markers of regional atrophy patterns
Multimodal approaches combining structural and molecular imaging
Blood-based biomarkers:
Plasma or serum TAF15 or fragments
Exosomal TAF15 from CNS origin
RNA profiles reflecting TAF15 dysfunction
Inflammatory markers associated with TAF15 pathology
Methodological development needs:
Ultra-sensitive assays for TAF15 detection (e.g., SIMOA)
TAF15 filament-specific antibodies
TAF15 seed amplification assays (similar to α-synuclein PMCA/RT-QuIC)
Longitudinal studies correlating biomarker changes with clinical progression
The development of such biomarkers would enable earlier diagnosis, monitoring of disease progression, and evaluation of therapeutic efficacy in clinical trials targeting TAF15 pathology.
TAF15 is a member of the FET (FUS, EWS, TAF15) family of proteins, which are structurally similar and multifunctional . These proteins were initially discovered due to their involvement in fusion oncogenes in human sarcomas and leukemias . TAF15 contains RNA recognition motifs (RRMs) and glycine-rich regions, which enable it to bind RNA and DNA .
The primary function of TAF15 is to facilitate the assembly of the transcription complex by interacting with other transcription factors and coactivators . It plays a crucial role in the regulation of gene expression by binding to the core promoter region of genes and positioning RNA polymerase II correctly . Additionally, TAF15 is involved in various cellular processes, including RNA splicing, transport, and stability .
Recombinant TAF15 is a form of the protein that is produced using recombinant DNA technology. This involves inserting the TAF15 gene into an expression vector, which is then introduced into a host cell (such as E. coli) to produce the protein . The recombinant protein can be purified and used for various research applications, including studying its function, interactions, and role in diseases .
The recombinant TAF15 protein typically includes a His-tag at the N-terminus to facilitate purification . It corresponds to the amino acids 148-406 of the human TAF15 protein .
TAF15 has been implicated in several diseases, particularly cancers. Translocations involving the TAF15 gene and other genes, such as the nuclear receptor CSMF, result in fusion proteins that are associated with a subset of extraskeletal myxoid chondrosarcomas . These fusion proteins can disrupt normal cellular functions and contribute to tumorigenesis .
Furthermore, TAF15 is involved in the regulation of transcription by RNA polymerase II, which is a critical process in maintaining cellular homeostasis and responding to various signals . Dysregulation of this process can lead to various diseases, including cancer .