Recombinant Human Transcription initiation factor TFIID subunit 8 (TAF8)

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference in order notes for customized preparation.
Lead Time
Delivery times vary depending on purchasing method and location. Contact your local distributor for precise delivery estimates.
Note: Standard shipping includes blue ice packs. Dry ice shipping requires prior arrangement and incurs additional charges.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to consolidate the contents. Reconstitute the protein in sterile deionized water to a concentration of 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting at -20°C/-80°C. Our standard glycerol concentration is 50%, but this can be adjusted as needed.
Shelf Life
Shelf life depends on storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized forms have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt. Aliquot for multiple uses to prevent repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing. Specify your desired tag type during ordering for preferential development.
Synonyms
FLJ32821; hTAFII43; Protein taube nuss; TAF(II)43; taf8; TAF8 RNA polymerase II, TATA box binding protein (TBP) associated factor, 43kDa; TAF8_HUMAN; TAFII-43; TAFII43; TATA box binding protein (TBP) associated factor, RNA polymerase II A 45/50kDa; Taube nuss homolog (mouse); TBN; TBP associated factor 8; TBP associated factor TAFII43; TBP associated factor, RNA polymerase II, 43 kD; TBP-associated factor 43 kDa; TBP-associated factor 8; Transcription initiation factor TFIID 43 kDa subunit; Transcription initiation factor TFIID subunit 8
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
2-310
Protein Length
Full Length of Mature Protein
Purity
>85% (SDS-PAGE)
Species
Homo sapiens (Human)
Target Names
TAF8
Target Protein Sequence
ADAAATAGA GGSGTRSGSK QSTNPADNYH LARRRTLQVV VSSLLTEAGF ESAEKASVET LTEMLQSYIS EIGRSAKSYC EHTARTQPTL SDIVVTLVEM GFNVDTLPAY AKRSQRMVIT APPVTNQPVT PKALTAGQNR PHPPHIPSHF PEFPDPHTYI KTPTYREPVS DYQVLREKAA SQRRDVERAL TRFMAKTGET QSLFKDDVST FPLIAARPFT IPYLTALLPS ELEMQQMEET DSSEQDEQTD TENLALHISM EDSGAEKENT SVLQQNPSLS GSRNGEENII DNPYLRPVKK PKIRRKKSLS
Uniprot No.

Target Background

Function
Transcription factor TFIID is a general factor essential for accurate and regulated RNA polymerase II-mediated transcription initiation. It facilitates both basal and activator-dependent transcription. While implicated in preadipocyte fibroblast differentiation into adipocytes, its role in myoblast differentiation appears minimal. It's crucial for TAF10 integration into the TAF complex and may be vital for the survival of inner cell mass cells, which comprise the early embryo's pluripotent cell population.
Gene References Into Functions
  1. Analysis of the TAF2-TAF8-TAF10 complex reveals a stepwise assembly pathway for nuclear holo-TFIID, regulated by nuclear import of preformed cytoplasmic submodules. PMID: 25586196
Database Links

HGNC: 17300

OMIM: 609514

KEGG: hsa:129685

STRING: 9606.ENSP00000362068

UniGene: Hs.520122

Protein Families
TAF8 family
Subcellular Location
Nucleus. Cytoplasm.

Q&A

What is TAF8 and what is its role in transcription?

Transcription initiation factor TFIID subunit 8 (TAF8) is a key component of the general transcription factor TFIID, which plays a fundamental role in RNA polymerase II (Pol II) transcription initiation in eukaryotic cells. TFIID is a megadalton-sized multiprotein complex composed of the TATA-binding protein (TBP) and 13 TBP-associated factors (TAFs) . TAF8 specifically contributes to the recognition of core promoter sequences and neighboring chromatin marks, and can participate in interactions with gene-specific activators and repressors . It serves as a critical structural component that helps maintain the integrity and functionality of the TFIID complex, which is essential for proper transcription initiation at most Pol II-dependent promoters.

How does TAF8 contribute to TFIID assembly?

TAF8 plays a central role in nucleating the TFIID complex. Research has revealed that TAF8 forms a heterotrimeric complex with TAF2 and TAF10 in the cytoplasm, known as the cTAF complex . This cytoplasmic TAF2-TAF8-TAF10 complex is a critical intermediate in TFIID assembly. TAF8 interacts with TAF10 via their histone fold domains (HFDs) in a non-canonical arrangement . Additionally, TAF8 binds to multiple motifs within the TAF2 protein through its C-terminal region . These interactions position TAF8 as a central nucleating factor that helps orchestrate the proper assembly of the complete TFIID complex. The TAF8 protein contains a nuclear localization sequence (NLS) that allows the cTAF complex to enter the nucleus, where TFIID assembly continues .

What is the structural organization of TAF8?

TAF8 contains a histone fold domain (HFD) that forms a heterodimer with the HFD of TAF10, creating a non-canonical histone pair arrangement . X-ray crystallography has revealed that both TAF8 and TAF10 have similar L1 loop geometries that are not found in other structures of related HFD-containing TAFs. In both proteins, a phenylalanine in loop L1 (F50 in TAF8 and F144 in TAF10) is embedded in a composite hydrophobic cavity mainly formed by residues from helices α1/α2 of one protein and helices α2/α3 of the other . Beyond its HFD, TAF8 also has a C-terminal region that contains multiple motifs important for interaction with TAF2 . Additionally, an extension from the HFD of TAF8 interacts with the homodimer of TAF6 HEAT repeats and connects it to the TAF2 subunit . This structural arrangement places TAF8 in a position to serve as a key connector between different components of the TFIID complex.

How does TAF8 deficiency affect organism development?

TAF8 deficiency has profound developmental consequences across multiple model systems. In mice, complete knockout of Taf8 is lethal very early in development, around embryonic day 4.0 (E4.0), due to apoptosis in the inner cell mass (ICM) of blastocysts . Conditional deletion of Taf8 in mouse embryonic stem cells (mESCs) leads to massive cell death after 8 days of treatment . Interestingly, when Taf8 is specifically deleted in the developing mouse central nervous system, apoptosis is unexpectedly restricted to forebrain regions . This selective vulnerability suggests tissue-specific requirements for TAF8 function.

The developmental importance of TAF8 is further underscored by a case study of a patient with a homozygous TAF8 mutation who presented with intellectual disability, developmental delay, and mild microcephaly . This mutation (c.781–1G > A) generated a frame shift resulting in an unstable TAF8 mutant protein with an unrelated C-terminus that was undetectable in the patient's cells . These findings collectively demonstrate that TAF8 plays critical roles in early development, with particularly important functions in neural development and cognitive function.

What is the relationship between TAF8 and p53-mediated apoptosis?

TAF8 deficiency triggers p53-mediated apoptotic cell death, particularly in neural tissues. Research in mouse models with conditional Taf8 deletion in the developing central nervous system has revealed that the absence of TAF8 leads to elevated nuclear levels of the transcription factor p53 . This increase in p53 is accompanied by upregulation of pro-apoptotic p53 target genes including Noxa, Puma, and Bax, which induce programmed cell death .

What are the proposed mechanisms for how TAF8 regulates transcription?

TAF8 appears to regulate transcription through multiple mechanisms. As a component of TFIID, TAF8 contributes to core promoter recognition and transcription initiation by RNA polymerase II . Recent studies suggest that TAF8 specifically supports the directionality of transcription and co-transcriptional splicing . When Taf8 is deleted in mice, aberrant transcription of promoter regions and splicing anomalies are observed .

Within the TFIID complex, TAF8 helps maintain the proper structural conformation needed for promoter recognition. The TFIID complex undergoes substantial conformational changes during promoter binding, with TBP and TFIIA engaging the TATA-end of the promoter after detaching from one part of the complex (Lobe A) and engaging with another part (Lobe B) . TAF8, along with other TAFs, forms part of the structure that interacts with specific promoter elements, including the motif ten element (MTE), downstream promoter element (DPE), and initiator (Inr) sequences .

Furthermore, TAF8 plays a role in establishing the asymmetry of the TFIID complex. The incorporation of the TAF8/TAF10 pair into a preexisting TAF subcomplex may break potential symmetry and allow the incorporation of other TAF subunits, thus generating the distinct lobes observed in the final TFIID structure . This structural contribution is likely critical for proper TFIID function in gene expression.

What methods can be used to study TAF8 protein-protein interactions within TFIID?

Several complementary approaches have proven effective for investigating TAF8's interactions within TFIID:

  • Immunoprecipitation and co-immunoprecipitation: Researchers have used antibodies against TAF8 or its interaction partners (like TAF2, TAF10, TAF7, TBP) to precipitate protein complexes from cell extracts and analyze the co-precipitated proteins by western blotting . This approach has been instrumental in identifying TAF8-containing complexes from different cellular compartments, including the cytoplasmic TAF2-8-10 complex .

  • Native mass spectrometry: This technique has been used to define the interactions between TAFs and uncover TAF8's central role in nucleating the complex . Mass spectrometry provides high-resolution data on protein complex composition and can detect both stable and transient interactions.

  • X-ray crystallography: This method has revealed the three-dimensional structure of the TAF8-TAF10 histone fold domain pair, showing their non-canonical arrangement . Such structural information is crucial for understanding the molecular basis of protein-protein interactions.

  • Chemical crosslinking-mass spectrometry (CX-MS): This approach has been used alongside cryo-electron microscopy to generate models of parts of TFIID, including regions containing TAF8 . Crosslinking captures protein-protein interactions in their native state before mass spectrometric analysis.

  • Recombinant protein expression and in vitro binding assays: Expressing TAF8 and potential binding partners in systems like baculovirus-infected Sf9 insect cells allows for controlled studies of direct interactions . FLAG-tagged versions of TAF8 have been used to facilitate purification and detection.

These methods can be combined to build a comprehensive understanding of how TAF8 interacts with other TFIID components in different cellular contexts.

How can researchers generate and validate TAF8 knockout or conditional knockout models?

Generating and validating TAF8 knockout or conditional knockout models requires careful experimental design due to the essential nature of this gene. Several approaches have been documented:

  • Conditional knockout strategy: Given that complete Taf8 knockout is embryonic lethal , conditional systems are necessary. The Cre-loxP system has been successfully employed, with Taf8 lox/lox mice crossed with tissue-specific Cre driver lines (like CNS-specific Cre lines) for targeted deletion .

  • Inducible systems: Taf8 lox/lox: Rosa26-CreERT2 mouse embryonic stem cells have been created, in which Taf8 deletion can be induced by the addition of 4-hydroxy Tamoxifen (4-OH) . This allows for temporal control of gene deletion.

  • Validation approaches:

    • mRNA analysis: RT-PCR or RNA-seq to confirm reduction in Taf8 transcript levels .

    • Protein analysis: Western blotting with anti-TAF8 antibodies to verify protein loss .

    • Phenotypic analysis: For embryonic models, assessment of development, apoptosis (using markers like cleaved caspase-3), and cell viability .

    • Functional validation: Analyzing the composition of TAF-containing complexes by immunoprecipitation with antibodies against other TFIID components (TAF7, TBP, TAF10) to demonstrate altered complex formation .

  • Rescue experiments: Complementation with wild-type TAF8 can confirm that phenotypes are specifically due to TAF8 loss. This approach has been used in TAF10 null mouse F9 cells with TAF10 HFD and chimeric mutants .

  • Double knockout approaches: Creating double knockouts (e.g., Taf8 and p53) can help dissect the pathways downstream of TAF8 loss, as demonstrated by the rescue of apoptosis but not the transcriptional defects in Taf8/p53 double knockout mice .

These models provide powerful tools for understanding TAF8's functions in different cellular contexts and developmental stages.

What techniques are available for analyzing the stability of wild-type versus mutant TAF8 proteins?

Several complementary techniques have been employed to analyze the stability of wild-type versus mutant TAF8 proteins:

  • Western blot analysis: This fundamental technique can detect and quantify TAF8 protein levels in cell extracts. Comparisons between wild-type and mutant samples reveal differences in steady-state protein abundance . In the case of the patient with TAF8 mutation (c.781–1G > A), western blotting showed undetectable levels of the mutant protein despite normal mRNA levels .

  • Recombinant protein expression systems: Expressing wild-type and mutant TAF8 proteins in heterologous systems (like baculovirus-infected Sf9 insect cells) allows for controlled comparison of protein stability . N-terminal epitope tagging (e.g., FLAG tag) facilitates detection and purification of recombinant proteins.

  • Proteasome inhibition experiments: Treating cells with proteasome inhibitors (e.g., MG132) can reveal whether protein degradation occurs via the ubiquitin-proteasome pathway. If protein levels increase after inhibitor treatment, this suggests proteasomal degradation is responsible for the reduced stability .

  • Pulse-chase experiments: This approach allows for tracking the fate of newly synthesized proteins over time. Cells are briefly exposed to radioactively labeled amino acids (pulse), followed by incubation in non-radioactive medium (chase). The labeled proteins are then analyzed at various time points to determine their half-lives .

  • mRNA level analysis: Quantifying mRNA levels (e.g., by RT-PCR or RNA-seq) alongside protein analysis helps distinguish between transcriptional and post-transcriptional effects. In the case of the patient with TAF8 mutation, mRNA levels were comparable between control and patient cells, indicating that protein instability rather than reduced transcription was responsible for the absence of detectable TAF8 protein .

These methods collectively provide a comprehensive assessment of protein stability and the mechanisms responsible for any observed differences between wild-type and mutant TAF8 proteins.

How do researchers reconcile the differential effects of TAF8 loss in different cell types?

The different effects of TAF8 loss across various cell types present an intriguing research puzzle. Several hypotheses have been proposed to explain these cell type-specific responses:

  • Differential dependency on canonical TFIID: The varying effects of TAF8 loss suggest that different cell types may have varying requirements for intact TFIID complexes. While embryonic stem cells and blastocysts show severe phenotypes including cell death and global transcription decreases upon TAF8 loss , patient fibroblasts with undetectable TAF8 show surprisingly minimal effects on global Pol II transcription . This suggests that some cell types may utilize alternative transcription initiation mechanisms that can compensate for canonical TFIID deficiency.

  • Partial TAF complexes: Research has shown that in TAF8-deficient cells, partial TAF complexes still exist . These partial complexes, or perhaps an altered TFIID containing trace amounts of mutated TAF8, might support sufficient transcription for survival in certain cell types but not others . The functional capacity of these partial complexes likely varies by cell type and developmental stage.

  • Developmental context: In mouse models, the trophectoderm cells of Taf8-deficient blastocysts remained intact in vivo and could be cultured in vitro for at least 10 days, while inner cell mass cells underwent apoptosis . Similarly, conditional TAF8 deletion in the developing mouse CNS caused apoptosis specifically in forebrain regions . These observations suggest that the developmental context and differentiation state significantly influence a cell's response to TAF8 deficiency.

  • Compensatory mechanisms: Different cell types may have varying capacities to upregulate compensatory pathways when TFIID function is compromised. These might include alternate transcription initiation complexes or cell-specific survival pathways that buffer against transcriptional disturbances.

  • Threshold effects: The requirement for TAF8 may involve threshold effects where certain cell types require higher levels of TFIID function than others to maintain viability and proper function.

These differential responses highlight the complexity of transcriptional regulation across cell types and developmental stages, and provide important insights into the context-dependent functions of general transcription factors.

What contradictions exist in the current understanding of TAF8's role in TFIID assembly?

Several contradictions have emerged in the literature regarding TAF8's role in TFIID assembly:

These contradictions highlight the complexity of TFIID assembly and function, and indicate that our understanding of this critical transcription factor complex is still evolving.

How does current research address the discrepancy between TAF8 mutation effects in human patients versus mouse models?

The striking discrepancy between the effects of TAF8 deficiency in human patient cells versus mouse models has prompted several explanations:

  • Species-specific differences: The most fundamental explanation may be species-specific differences in transcriptional machinery requirements. Human cells may have evolved redundancies or compensatory mechanisms not present in mice that allow them to maintain transcription despite TAF8 deficiency .

  • Cell type specificity: The cell types examined in the different models may have intrinsically different requirements for TAF8. Patient fibroblasts showing minimal transcriptional changes may be less dependent on canonical TFIID than mouse embryonic stem cells or neuronal cells that show severe phenotypes upon TAF8 loss .

  • Complete vs. partial loss: The mouse models typically involve complete deletion of Taf8 , while the human patient had a mutation (c.781–1G > A) that generated an unstable protein with an altered C-terminus . Though the mutant protein was undetectable by standard methods, trace amounts may retain partial function or form partial complexes that support basic transcription.

  • Developmental timing: The effects of TAF8 loss may be particularly severe during specific developmental windows. Mouse studies often examine embryonic stages or embryonic stem cells , while the human patient studies examined differentiated fibroblasts . The timing of TAF8 loss may significantly influence the observed phenotypes.

  • Gene dosage effects: The degree of TAF8 deficiency might differ between models. Even minimal residual TAF8 function in the human cells might be sufficient to prevent the severe effects seen in complete knockout models.

  • Methodological differences: The assays used to measure transcriptional effects (e.g., RNA-seq, Pol II ChIP-seq) have technical limitations and might not detect subtle but biologically significant changes in specific gene sets or transcription processes.

Researchers are addressing these discrepancies through more sophisticated models, including:

  • Creating human cellular models with complete TAF8 knockout using CRISPR-Cas9

  • Generating knock-in models of patient-specific mutations in mice

  • Developing tissue-specific and inducible Taf8 deletion models to better control the timing and extent of TAF8 loss

  • Performing more detailed analyses of transcriptional fidelity, including assessment of transcription directionality, splicing accuracy, and promoter specificity

These approaches will help clarify whether the observed differences reflect genuine biological distinctions or technical limitations of current experimental models.

What are the emerging therapeutic implications of TAF8 research for neurodevelopmental disorders?

The discovery of TAF8 mutations in patients with intellectual disability and developmental delay has opened new avenues for understanding and potentially treating neurodevelopmental disorders:

These therapeutic directions, while still largely theoretical, represent promising avenues for translating basic TAF8 research into clinical applications for neurodevelopmental disorders.

What novel techniques are being developed to study TFIID assembly and function in living cells?

Researchers are developing several cutting-edge techniques to study TFIID assembly and function in living cells:

  • Live-cell imaging of fluorescently tagged TAF proteins: Advanced fluorescent protein tagging combined with super-resolution microscopy allows visualization of TFIID component dynamics in living cells. Techniques like FRAP (Fluorescence Recovery After Photobleaching) can measure the exchange rates of individual TAFs within complexes, providing insights into TFIID stability and assembly kinetics.

  • Single-molecule tracking: This approach allows researchers to follow individual TFIID complexes in real time, revealing the dynamics of complex assembly, promoter searching, and engagement with transcriptional machinery. By labeling different TAF subunits, including TAF8, researchers can determine the order and kinetics of complex assembly.

  • Proximity labeling techniques: Methods like BioID or APEX2 allow researchers to identify proteins in close proximity to a protein of interest (e.g., TAF8) in living cells. These approaches can capture transient interactions during TFIID assembly that might be missed by traditional immunoprecipitation methods.

  • CRISPR-based genomic tagging: CRISPR-Cas9 technology enables precise tagging of endogenous TAF genes with fluorescent proteins or affinity tags, allowing visualization or purification of TAF proteins expressed at physiological levels from their native genomic contexts.

  • Optogenetic control of TFIID assembly: Light-inducible protein interaction domains can be engineered into TAF proteins to control their assembly into complexes with spatial and temporal precision, enabling detailed studies of assembly pathways and the consequences of specific complex formations.

  • Single-cell transcriptomics combined with TFIID component perturbation: This approach allows researchers to correlate variations in TFIID composition with transcriptional outputs at the single-cell level, revealing the functional consequences of different TFIID assembly states.

  • Cryo-electron tomography of intact cells: This emerging technique may eventually allow visualization of TFIID complexes in their native cellular environment, providing insights into their organization and interactions that are not apparent in purified systems.

These advanced techniques promise to provide unprecedented insights into the dynamic assembly and function of TFIID in living cells, potentially resolving current contradictions and revealing new principles of transcriptional regulation.

How might understanding TAF8's role in transcriptional fidelity inform broader questions about gene regulation?

Understanding TAF8's role in transcriptional fidelity has significant implications for broader concepts in gene regulation:

  • Transcriptional directionality control: Research suggests that TAF8 supports the directionality of transcription . This finding connects to the emerging understanding that promoters are inherently bidirectional, and specific factors are required to enforce directional transcription. TAF8 research may reveal fundamental mechanisms by which cells ensure proper transcriptional orientation, with implications for understanding pervasive non-coding transcription and its regulation.

  • Co-transcriptional processing coordination: TAF8 deficiency causes splicing anomalies , highlighting the intimate connection between transcription initiation and RNA processing. Further investigation may reveal how components of the basal transcription machinery influence downstream RNA processing events, potentially through controlling RNA polymerase II elongation rates or recruiting processing factors.

  • Cell type-specific transcription requirements: The differential effects of TAF8 loss across cell types contribute to our understanding of how apparently "general" transcription factors may have specialized roles in different cellular contexts. This concept challenges the traditional distinction between general and specific transcription factors and suggests a more nuanced view of transcriptional regulation across cell types.

  • Transcriptional robustness and compensatory mechanisms: The surprising finding that human patient cells lacking detectable TAF8 show minimal transcriptional defects reveals previously unappreciated robustness in the transcription system. Understanding how cells maintain transcription despite deficiencies in core machinery components may uncover important backup systems that ensure gene expression fidelity under stress conditions.

  • Evolution of transcriptional complexity: The complex architecture of TFIID, with the pseudo-duplication of core components in different lobes , raises questions about the evolution of eukaryotic transcription systems. TAF8 research may provide insights into how increasingly complex transcription initiation machinery evolved to support the sophisticated gene regulatory networks in higher eukaryotes.

  • Transcriptomopathies as a disease paradigm: The concept that broad transcriptional disturbances can lead to specific disease phenotypes, particularly affecting the nervous system , represents an important paradigm shift in understanding genetic disorders. TAF8 research contributes to this emerging field of "transcriptomopathies" and may help explain why seemingly general transcriptional defects often manifest with tissue-specific phenotypes.

By elucidating these broader principles, TAF8 research extends beyond understanding a single protein to informing fundamental concepts in gene regulation that have implications across biology and medicine.

What expression systems are most effective for producing functional recombinant TAF8 protein?

Based on the research literature, several expression systems have been employed for producing recombinant TAF8, each with distinct advantages for different research applications:

  • Baculovirus-infected Sf9 insect cells: This system has been successfully used to express both wild-type and mutant TAF8 proteins, particularly with N-terminal epitope tags like FLAG . The baculovirus system is advantageous for expressing eukaryotic proteins as it provides post-translational modifications and appropriate protein folding machinery. It has been particularly useful for studying TAF8 stability and interactions with other TFIID components.

  • Bacterial expression systems: While not explicitly mentioned in the provided search results, E. coli systems are commonly used for expressing protein domains for structural studies. For TAF8, this approach might be suitable for expressing the histone fold domain for crystallography studies, as was likely done for the TAF8-TAF10 HFD structural analysis .

  • Mammalian expression systems: For studies requiring the most physiologically relevant post-translational modifications and folding, mammalian cell expression (e.g., HEK293 cells) would be appropriate, especially when studying TAF8's interactions with other human proteins or its incorporation into partial or complete TFIID complexes.

When producing recombinant TAF8, several technical considerations are important:

  • Co-expression strategies: Given TAF8's tendency to form complexes with other TAFs, co-expressing TAF8 with partners like TAF10 may improve solubility and stability. This approach is particularly relevant for structural studies of TAF subcomplexes.

  • Epitope tagging: N-terminal tags (FLAG, His, GST) have been successfully used with TAF8 . The position of the tag is important, as C-terminal tags might interfere with interactions mediated by the C-terminal region of TAF8 that are critical for binding to TAF2 .

  • Protein stability considerations: The stability issues observed with the patient-derived mutant TAF8 (with altered C-terminus) highlight the importance of optimizing expression and purification conditions to maintain protein stability. This may include using protease inhibitors and optimizing buffer conditions.

  • Functional validation: Assessing the functionality of recombinant TAF8 is crucial, particularly when using it for interaction studies or in vitro reconstitution of TFIID subcomplexes. Methods like the complementation assays used for TAF10 in TAF10 null mouse F9 cells could be adapted for TAF8.

The choice of expression system should be guided by the specific research question, with insect cell systems offering a good balance of yield and physiological relevance for most TAF8 applications in biochemical and structural studies.

What bioinformatic approaches are recommended for analyzing TAF8-dependent transcriptional changes?

For comprehensive analysis of TAF8-dependent transcriptional changes, researchers should consider multiple bioinformatic approaches:

These bioinformatic approaches should ideally be combined in a comprehensive analysis pipeline to fully characterize the complex transcriptional changes resulting from TAF8 deficiency or mutation.

How does TAF8 function compare with other TAF proteins in the TFIID complex?

TAF8 possesses both shared and unique characteristics compared to other TAF proteins within the TFIID complex:

This comparative analysis highlights TAF8's distinctive role as a central organizer within TFIID, with unique structural features and assembly functions that differentiate it from other TAFs despite sharing the common HFD structural motif found in many TFIID components.

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 2025 TheBiotek. All Rights Reserved.