THAP11 acts as a transcriptional repressor by recruiting chromatin-modifying complexes. Key findings include:
DNA Binding: Binds promoter regions (e.g., c-Myc) via its THAP domain, recruiting histone deacetylases (HDACs) to repress transcription .
HCF-1 Interaction: Forms a complex with HCF-1 (Host Cell Factor-1) to regulate genes involved in cell cycle and metabolism .
c-Myc Repression: Overexpression reduces c-Myc protein levels, inhibiting cell proliferation .
A homozygous F80L mutation in THAP11 disrupts its interaction with HCF-1, impairing MMACHC gene regulation and causing cobalamin deficiency. This mirrors cblX disorder but without HCFC1 mutations .
Cobalamin Disorder Model:
Cancer Models:
THAP11 is a member of the Thanatos-associated protein (THAP) family, which consists of zinc-dependent, sequence-specific DNA-binding factors involved in cell proliferation, apoptosis, cell cycle regulation, chromatin modification, and transcriptional regulation . The protein contains a highly conserved THAP zinc-finger domain at the N-terminus that enables sequence-specific DNA binding activity . Additionally, the protein structure features a 29-residue repeat polyglutamine motif basic domain in the middle of the sequence and a weak nuclear localization signal in the region from amino acids 98 to 103 .
The functional importance of THAP11's structure has been confirmed through localization studies. When THAP11 cDNA is fused to EGFP and transfected into cells, the resulting fusion protein demonstrates clear nuclear localization, confirming that the nuclear localization signal in the protein is indeed functional . This nuclear localization is critical for THAP11's role as a transcriptional regulator, allowing it to interact directly with DNA in the nucleus.
THAP11 is encoded by a gene located on chromosome 16q22.1 and has been suggested as a potential candidate for polyglutamine disorders based on polymorphism and protein-folding simulation studies . The human THAP11 protein shares significant homology with its mouse counterpart, called Ronin, particularly in the DNA-binding THAP domain region .
Analysis using Cancer Profiling Array II has shown that THAP11 is frequently downregulated in several types of human tumor tissues compared to their corresponding normal tissues. Specifically, downregulation has been observed in tumor samples derived from the liver (100% of samples), thyroid gland (70%), vulva (60%), skin (50%), pancreas (approximately 57%), and kidney (40%) . In contrast, THAP11 shows significant upregulation in tumor samples from the lung (70%) and ovary (40%) .
This varied expression pattern suggests that THAP11 may play different roles in different types of cancers, potentially functioning as a tumor suppressor in some tissues while possibly contributing to oncogenesis in others. The mechanism behind this tissue-specific differential expression remains an area requiring further investigation, as it may provide insights into tissue-specific cancer pathogenesis and potential therapeutic targets.
THAP11 functions as a potent growth suppressor in various cell types through its ability to repress transcription of the c-Myc oncogene . This regulatory mechanism has been demonstrated through multiple experimental approaches. When THAP11 is overexpressed in human hepatoma cell lines (7721), the proliferation rate significantly decreases compared to control cells . Colony formation assays further confirm this effect, showing approximately 60% reduction in colony formation in cells expressing THAP11 .
The growth suppression effect of THAP11 has been validated through RNA interference experiments. When endogenous THAP11 expression is knocked down in HepG2 cells using siRNA (with 65-84% efficiency at the mRNA level and 53-70% at the protein level), an increased proliferation rate is observed compared to control cells . This indicates that THAP11 serves as an endogenous physiologic regulator of cell proliferation.
The growth suppression effect of THAP11 is not limited to hepatocellular carcinoma cells but extends to various cell types including human diploid fibroblasts (2BS), human cervix adenocarcinoma cells (HeLa), and human breast cancer cells (MCF-7) . In MTT assays with human fibroblast cells, THAP11 overexpression decreased proliferation rates by approximately 40% . This broad-spectrum growth suppression activity suggests that THAP11 regulates fundamental aspects of cell proliferation that are conserved across different cell types.
THAP11 has been identified as a recently discovered pluripotency factor involved in embryonic stem cell (ESC) self-renewal and embryonic development . It appears to function independently of other known pluripotency factors, suggesting a unique mechanism of action in maintaining stem cell properties .
Experimental evidence indicates that overexpression of THAP11 in human fibroblast cells leads to increased expression levels of key pluripotency genes including Sox2, Oct4, Nanog, and Klf4 . This finding is particularly significant as these four factors are essential components of the core pluripotency network that maintains the undifferentiated state of stem cells and controls their capacity for self-renewal.
In mouse studies, knockout of the THAP11 gene (called Ronin in mice) proved lethal during early embryonic development, while overexpression in ESCs prevented differentiation and promoted teratocarcinoma formation . This dual functionality - simultaneously acting as a pluripotency factor and a proliferation suppressor - represents a complex molecular mechanism that remains to be fully elucidated.
The mechanism by which THAP11 regulates pluripotency appears to involve its DNA-binding activity. THAP11 contains an evolutionarily conserved zinc finger motif (the THAP domain) with sequence-specific DNA-binding capabilities . Research suggests that Ronin (mouse THAP11) may regulate cell cycle controller genes by binding to hyperconserved shared enhancers, potentially through interactions with other factors including HCF1 and ZNF143 .
THAP11 exerts its growth suppression effects primarily through the selective inhibition of c-Myc expression, without affecting other proliferation-related genes such as c-jun and c-fos . This selective repression occurs at the transcriptional level through direct binding of THAP11 to the c-Myc promoter region.
The molecular mechanism has been elucidated through several experimental approaches. In inducible THAP11 expression systems (293-THAP11 cells), treatment with Ponasterone A (PonA) to induce THAP11 expression resulted in approximately 50% reduction in c-Myc protein levels after 36 hours . Conversely, when endogenous THAP11 was knocked down in HepG2 cells using siRNA, c-Myc expression increased 2-2.5 fold .
Reporter assays using c-Myc promoter constructs demonstrate that THAP11 represses c-Myc promoter activity in a dose-dependent manner . In 293-THAP11 cells, induction with 5 μM PonA for 24 hours reduced c-Myc promoter activity by approximately 52% . Similar repression was observed in MCF-7, HeLa, and HepG2 cells when THAP11 was co-transfected with the c-Myc promoter reporter construct .
Chromatin immunoprecipitation (ChIP) assays have revealed that THAP11 binds specifically to the c-Myc promoter in vivo, with binding restricted to the region between -710 to -463 bp from the transcription start site . Treatment with PonA resulted in high occupancy by THAP11 at this region, showing an approximately 30-fold increase compared to control conditions . In contrast, no significant binding was observed at the -1218 to -918 bp region, indicating a specific interaction with the -710 to -463 bp promoter region .
The dual functionality of THAP11 as both a pluripotency factor and a growth suppressor represents a fascinating paradox in cellular biology, as these two functions appear contradictory. Pluripotency is typically associated with proliferative capacity, while growth suppression inherently limits cell division.
Experimental evidence clearly demonstrates this dual nature. Overexpression of THAP11 in human fibroblast cells simultaneously increases the expression of key pluripotency genes (Sox2, Oct4, Nanog, and Klf4) while decreasing proliferation rates by approximately 40% . This confirms that researchers are "faced with a molecule with double features, which could be involved in pluripotency and proliferation suppressor simultaneously" .
Several possibilities might explain this duality:
Context-dependent regulation: THAP11 may interact with different cofactors in different cellular contexts, leading to distinct gene regulation patterns
Feedback mechanisms: THAP11 might initiate pluripotency programs that subsequently activate negative feedback loops to control proliferation
Complex interplay with other regulators: The balance between pluripotency and growth suppression might depend on interactions with other cellular factors that modulate THAP11 activity
The researchers conclude that "the roles of THAP11 in pluripotency are so complex and attributed to other regulatory molecules" , suggesting that unraveling this dual functionality requires investigation of the broader regulatory network around THAP11.
Studying THAP11's DNA binding specificity requires a comprehensive experimental approach combining in vitro and in vivo techniques. Based on the research methodologies described in the available literature, the following approaches are recommended:
Chromatin Immunoprecipitation (ChIP) assays have proven effective for studying THAP11's DNA binding in vivo . This technique revealed that THAP11 specifically binds to the c-Myc promoter region between -710 to -463 bp . For optimal results, ChIP assays should be coupled with quantitative PCR (qPCR) to measure binding enrichment at specific genomic regions compared to control regions. In the case of THAP11 binding to the c-Myc promoter, this approach demonstrated a 30-fold increase in occupancy at the target region .
Reporter gene assays provide functional validation of THAP11 binding sites. By constructing reporter plasmids containing putative THAP11 binding regions (as done with the c-Myc promoter), researchers can assess the functional impact of THAP11 binding on gene expression . These assays should include both wild-type sequences and mutated binding sites to confirm specificity. Including dose-dependent analyses (as performed with PonA-inducible systems) can provide quantitative insights into THAP11's regulatory potential .
For systematic identification of THAP11 binding sites across the genome, ChIP followed by next-generation sequencing (ChIP-seq) would be ideal, although this approach was not explicitly described in the provided search results. This global approach would enable identification of consensus binding motifs and potential co-regulatory elements.
In vitro DNA binding assays such as electrophoretic mobility shift assays (EMSAs) can complement in vivo techniques by determining direct binding affinities and sequence requirements. Purified recombinant THAP11 protein (focusing on the THAP domain) can be used with labeled DNA probes containing predicted binding sites.
The expression pattern of THAP11 shows notable differences between embryonic stem cells (ESCs) and differentiated adult tissues, though there appears to be some discrepancy in the literature regarding the extent of this difference.
In mouse studies, Dejosez et al. reported that Ronin (mouse THAP11) expression in adult tissue was negligible , suggesting that THAP11 expression is primarily associated with the embryonic/stem cell state. This aligns with the finding that knocking out Ronin proved lethal during early embryonic development , indicating an essential role in embryogenesis.
The apparent contradiction might be explained by several factors:
Species differences: The negligible expression reported by Dejosez et al. was in mouse tissues, while Zhu et al. reported on human tissues
Detection sensitivity: Different methodologies may have varying sensitivities for detecting low-abundance transcripts
Tissue-specific variation: THAP11 expression might vary substantially between different adult tissues
Developmental regulation: Expression levels might change during development and aging
The overexpression of THAP11 in differentiated fibroblast cells leads to increased expression of pluripotency factors (Sox2, Oct4, Nanog, and Klf4) , suggesting that THAP11 might play a role in cellular reprogramming. This indicates that the relationship between THAP11 expression and cellular differentiation state is complex and potentially bidirectional.
Based on the search results, researchers have successfully employed several complementary approaches to modulate THAP11 expression in various experimental systems. These methodologies can be categorized into overexpression, knockdown, and inducible expression systems.
For THAP11 overexpression, researchers have utilized plasmid-based transient transfection and lentiviral-mediated stable transduction. In the plasmid-based approach, the full-length cDNA coding human THAP11 was cloned into expression vectors (such as pcDNA3.1) and transfected into target cells . The efficiency of transfection was monitored through GFP co-transfection in parallel wells . For more stable and efficient expression, particularly in primary cells, lentiviral transduction has proven effective. This approach involved cloning the THAP11 gene into a lentiviral vector (CDH-CMV-MCS-EF1-GFP-T2A-Puro) and producing lentiviral particles in HEK293T cells using helper vectors (psPAX2 and pMD2.G) . The viral particles were then used to transduce human fibroblast cells, with successful transduction monitored via GFP expression and selection ensured through puromycin resistance .
For THAP11 knockdown, RNA interference using small interfering RNAs (siRNAs) has been successfully applied. To rule out off-target effects, two different siRNA oligos (siTHAP11-1 and siTHAP11-2) were used, resulting in 65-84% reduction in THAP11 mRNA levels and 53-70% reduction in protein levels within 72 hours after transfection . This approach was particularly effective in HepG2 cells and provided valuable insights into the effects of THAP11 depletion on cell proliferation and c-Myc expression .
For controlled expression studies, an inducible expression system was developed in HEK293 cells (designated 293-THAP11 cells) . This system employed Ponasterone A (PonA), an ecdysone analog, as an inducing reagent to control THAP11 expression. The system showed both dose-dependent and time-dependent induction of THAP11 expression, with protein levels detectable 24 hours after PonA addition and accumulating thereafter . This inducible system proved valuable for studying the effects of THAP11 on c-Myc expression and transcriptional regulation in a controlled manner.
Reliable detection and quantification of THAP11 expression requires a multi-faceted approach employing both nucleic acid and protein-level analyses. Based on the methodologies described in the search results, several complementary techniques have proven effective.
At the mRNA level, reverse transcription-PCR (RT-PCR) has been successfully used to examine THAP11 expression profiles in human adult tissues using Multiple Tissue cDNA . For more quantitative analysis, real-time PCR (qPCR) has been employed to measure changes in THAP11 expression levels following experimental manipulations such as siRNA knockdown, showing up to 84% reduction in THAP11 mRNA levels . This technique is particularly valuable for measuring relative changes in expression across different experimental conditions or tissue samples.
For larger-scale expression profiling, hybridization-based methods such as the Cancer Profiling Array II have been used to evaluate THAP11 expression patterns across multiple tumor types and their corresponding normal tissues . This approach allows for simultaneous analysis of multiple samples, facilitating comparative studies across different tissues and disease states.
At the protein level, Western blotting using specific antibodies has been effectively applied to detect and quantify THAP11 protein expression . In systems where epitope tagging is possible, such as the inducible 293-THAP11 cells, V5 epitope-specific antibodies have been used to identify clones expressing THAP11 upon induction . Western blotting has also been used to monitor the efficiency of siRNA-mediated knockdown, showing 53-70% reduction in THAP11 protein levels .
For subcellular localization studies, fluorescence microscopy using GFP fusion proteins has proven effective. THAP11 cDNA fused to EGFP in pEGFP vector and transfected into COS-7 cells demonstrated nuclear localization of the fusion protein, confirming the functionality of the nuclear localization signal in THAP11 .
Studying THAP11's role in pluripotency requires careful experimental design considerations to address its complex regulatory functions and dual nature as both a pluripotency factor and proliferation suppressor. Based on the available research, several critical considerations emerge:
Selection of appropriate cell models is paramount. Since THAP11's functions appear to be context-dependent, researchers should consider multiple cell types including embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs), various progenitor cells, and differentiated cells like fibroblasts . This allows for comparative analysis across the spectrum of cellular differentiation states. The choice between human and mouse systems is also important, as there may be species-specific differences in THAP11/Ronin function and expression patterns .
Expression modulation strategies should be carefully designed. Both gain-of-function (overexpression) and loss-of-function (knockdown/knockout) approaches should be employed to comprehensively understand THAP11's roles . For overexpression studies, considering the use of inducible systems allows for temporal control and dose-dependent analysis, which is particularly valuable given THAP11's growth suppression effects . For knockdown studies, using multiple siRNA sequences helps rule out off-target effects .
Comprehensive readout systems are essential. Given THAP11's dual functionality, experiments should simultaneously assess:
Pluripotency markers: Gene and protein expression of core pluripotency factors (Sox2, Oct4, Nanog, Klf4) as conducted in fibroblast studies
Proliferation metrics: Cell counting, colony formation assays, or MTT assays to measure growth effects
Differentiation capacity: Ability of cells to maintain undifferentiated state or differentiate into multiple lineages
Molecular mechanism investigations should incorporate both DNA binding studies (ChIP assays) and transcriptional regulation analyses (reporter assays) . These should examine not only pluripotency-associated genes but also growth-related genes like c-Myc to understand the interplay between these seemingly contradictory functions .
Time course experiments are particularly important given the temporal dynamics of pluripotency networks and differentiation processes. THAP11's effects may vary at different stages of stem cell self-renewal, commitment, and differentiation, necessitating analysis at multiple time points.
THAP11's differential expression pattern across various cancer types and its established role in regulating cell proliferation suggest significant potential implications for cancer biology and therapeutics. The current research indicates several promising avenues for translational applications.
THAP11 demonstrates a tumor-specific expression pattern that varies by tissue type. It is downregulated in several human tumor tissues including liver (100% of samples), thyroid gland (70%), vulva (60%), skin (50%), pancreas (approximately 57%), and kidney (40%) compared to corresponding normal tissues . Conversely, THAP11 shows significant upregulation in tumor samples from the lung (70%) and ovary (40%) . This differential expression pattern suggests that THAP11 may function as a tumor suppressor in some tissues while potentially contributing to oncogenesis in others, indicating context-dependent roles in cancer development.
The growth suppression function of THAP11 through c-Myc repression represents a particularly significant mechanism relevant to cancer therapeutics . c-Myc is a well-established oncogene frequently deregulated in human cancers, contributing to uncontrolled proliferation. THAP11's ability to directly bind to the c-Myc promoter and repress its expression by approximately 52% in reporter assays suggests potential utility in cancer intervention strategies . Notably, this repression appears to be selective for c-Myc, as THAP11 did not affect expression of other proliferation-related genes like c-jun and c-fos .
In specific cancer types where THAP11 is downregulated, such as hepatocellular carcinoma, restoration of THAP11 expression could potentially serve as a therapeutic strategy. Experiments in hepatoma cell lines (7721) showed that THAP11 overexpression significantly decreased proliferation rates and reduced colony formation by approximately 60% . Similar growth suppression effects were observed in cervical adenocarcinoma (HeLa) and breast cancer (MCF-7) cell lines, with colony formation reductions of approximately 80% and 75%, respectively .
THAP11's dual role as both a pluripotency factor and a regulator of cell proliferation positions it as a molecule of significant interest for cellular reprogramming and regenerative medicine applications. The available research suggests several potential implications in these fields.
THAP11 overexpression in human fibroblast cells leads to increased expression of key pluripotency genes including Sox2, Oct4, Nanog, and Klf4 . This finding is particularly relevant to cellular reprogramming, as these four factors are central to induced pluripotent stem cell (iPSC) generation. Oct4 and Sox2 are among Yamanaka's original reprogramming factors, while Nanog is a key marker of successfully reprogrammed cells. The ability of THAP11 to upregulate these factors in differentiated cells suggests it might enhance reprogramming efficiency or perhaps serve as an alternative factor in reprogramming cocktails.
The function of THAP11 in embryonic stem cell self-renewal and embryo development further supports its potential utility in stem cell maintenance for regenerative medicine applications . Mouse studies have shown that Ronin (mouse THAP11) knockout is lethal in early embryonic development, while its overexpression in ESCs prevented differentiation . This suggests that modulating THAP11 levels could potentially help maintain stem cells in an undifferentiated state during in vitro expansion, addressing a key challenge in regenerative medicine.
The proposed mechanism of THAP11 action in pluripotency, involving regulation of cell cycle controller genes through binding to hyperconserved shared enhancers , suggests that it might influence epigenetic reprogramming processes. This could potentially be exploited to enhance the efficiency or completeness of cellular reprogramming.
THAP11's essential role in embryonic development suggests it may be implicated in developmental disorders and congenital diseases, though direct evidence in the provided search results is limited. Several lines of evidence point to potentially significant roles in human developmental pathologies.
THAP11 has been mapped to chromosome 16q22.1 and suggested as a putative candidate for polyglutamine disorders based on polymorphism and protein-folding simulation studies . This is particularly relevant given that THAP11 contains a 29-residue repeat polyglutamine motif in its protein structure . Polyglutamine expansion disorders, such as Huntington's disease and several spinocerebellar ataxias, result from abnormal expansion of CAG repeats encoding glutamine. Variations in THAP11's polyglutamine region could potentially contribute to similar neurological disorders, though specific disease associations require further investigation.
The lethal phenotype observed in mouse Ronin (THAP11) knockout models during early embryonic development underscores the protein's critical role in embryogenesis. This suggests that severe mutations in human THAP11 might result in embryonic lethality, contributing to early pregnancy loss. Less severe mutations might lead to developmental abnormalities compatible with life but resulting in congenital disorders.
THAP11's involvement in pluripotency regulation and its impact on key pluripotency genes (Sox2, Oct4, Nanog, and Klf4) indicates a potential role in developmental pathways that, when disrupted, could lead to congenital abnormalities. Sox2 mutations in humans are associated with anophthalmia (absence of eyes), while Oct4 (POU5F1) is essential for early embryonic development and primordial germ cell formation. Disruption of THAP11's regulation of these factors could potentially contribute to related developmental disorders.
The growth suppression function of THAP11 through c-Myc repression suggests that its dysregulation might affect embryonic growth patterns. Abnormal embryonic growth is associated with various developmental disorders, from growth restrictions to overgrowth syndromes. THAP11 mutations that alter its growth regulatory function might contribute to such conditions.
Based on the methodologies described in the search results, several effective cloning strategies have been successfully employed for THAP11 expression constructs. These approaches can be adapted based on specific experimental requirements.
PCR amplification of the THAP11 gene using specific primers with added restriction enzyme sites has proven effective . For optimal results, high-fidelity DNA polymerases such as Phusion DNA polymerase should be used to minimize PCR-induced mutations . The primers should be designed with appropriate restriction enzyme sites (e.g., XbaI/EcoRI as used in the fibroblast study) that are compatible with the target vector but absent from the THAP11 sequence . The following PCR cycling conditions have been successfully used: initial denaturation at 98°C for 30 seconds, followed by 30 cycles of denaturation (98°C, 10 seconds), annealing (60°C, 30 seconds), and extension (72°C, 30 seconds), with a final extension at 72°C for 10 minutes .
For expression vector selection, several options have proven successful depending on experimental goals:
Standard mammalian expression vectors such as pcDNA3.1 for transient transfection studies
Lentiviral vectors like CDH-CMV-MCS-EF1-GFP-T2A-Puro for stable integration, particularly useful for primary cells
Inducible expression systems for controlled expression studies
The inclusion of reporter genes and selection markers facilitates monitoring and selection of successfully transfected/transduced cells. GFP has been used effectively as a reporter in eukaryotic hosts, allowing visual confirmation of transfection/transduction efficiency . Selection markers such as puromycin resistance genes enable selection of stably transduced cells .
For lentiviral production, the three-plasmid transfection system has been effectively employed, using the expression construct together with helper vectors (psPAX2 and pMD2.G) in HEK293T cells . This approach generates viral particles capable of transducing various cell types, including primary human fibroblasts .
For specialized applications, fusion constructs can be created. THAP11 has been successfully fused to the N-terminus of EGFP to study subcellular localization, confirming its nuclear localization . Similar fusion strategies could be employed for protein purification (using histidine or GST tags) or protein-protein interaction studies (using FRET-compatible fluorescent proteins).
Designing effective THAP11 knockdown experiments requires careful consideration of several methodological aspects to ensure robust and interpretable results. Based on the approaches described in the search results, the following considerations are critical:
Selection of appropriate siRNA sequences is fundamental to effective knockdown experiments. To rule out off-target effects, multiple siRNA oligos targeting different regions of the THAP11 mRNA should be employed, as demonstrated with siTHAP11-1 and siTHAP11-2 in the hepatocellular carcinoma studies . These siRNAs achieved knockdown efficiencies of up to 84% at the mRNA level and 70% at the protein level . Custom design of siRNAs should follow established guidelines for effective siRNA design, including appropriate GC content, absence of internal repeats, and targeting regions unique to THAP11.
Validation of knockdown efficiency must be performed at both mRNA and protein levels. mRNA levels can be quantified using RT-PCR or qPCR techniques as employed in the HepG2 cell studies . Protein levels should be assessed via Western blotting using specific antibodies against THAP11 or epitope tags if working with tagged constructs . Establishing a time course of knockdown efficiency is advisable, as the studies showed assessments at 72 hours post-transfection .
Appropriate control conditions are essential. Experiments should include non-targeting siRNA controls with similar chemical modifications and delivery methods to the THAP11-targeting siRNAs. This controls for non-specific effects of the transfection process and presence of exogenous RNA .
Cell type selection should consider endogenous THAP11 expression levels. The knockdown studies in HepG2 cells were effective because these cells express detectable levels of endogenous THAP11 . Preliminary assessment of THAP11 expression in candidate cell lines is advisable before initiating knockdown experiments.
Functional readouts should be carefully selected based on THAP11's known functions. Given THAP11's established roles in cell proliferation regulation and pluripotency, appropriate assays include:
Proliferation assays such as those used in HepG2 cells following THAP11 knockdown
Expression analysis of c-Myc, which showed 2-2.5 fold increases following THAP11 knockdown
Assessment of pluripotency marker expression (Sox2, Oct4, Nanog, Klf4)
Analyzing THAP11's interactions with target gene promoters requires a multi-faceted approach combining several complementary techniques. Based on the methodologies employed in the search results, the following strategies are recommended:
Chromatin Immunoprecipitation (ChIP) has proven highly effective for studying THAP11 binding to target promoters in vivo . This technique successfully demonstrated THAP11 binding to the c-Myc promoter, with binding restricted to the -710 to -463 bp region . For optimal results, ChIP should be performed with specific antibodies against THAP11 or epitope-tagged versions in appropriate expression systems. Quantitative PCR following ChIP allows precise measurement of enrichment at target regions, as evidenced by the 30-fold enrichment observed for THAP11 binding to the c-Myc promoter region compared to control regions .
Promoter-reporter assays provide functional validation of THAP11 binding sites. This approach was successfully used to demonstrate that THAP11 represses c-Myc promoter activity by approximately 52% in a dose-dependent manner . For comprehensive analysis, researchers should construct reporter plasmids containing various lengths and mutations of the target promoter to map the precise binding sites and their functional importance. Testing these constructs in multiple cell types (as done with MCF-7, HeLa, and HepG2 cells) can reveal cell type-specific effects .
Inducible expression systems, such as the PonA-inducible system used for THAP11, allow for temporal control and dose-response studies of THAP11's effects on target promoters . This approach revealed that THAP11 repression of the c-Myc promoter was dose-dependent, providing insights into the quantitative aspects of this regulatory interaction .
For identifying THAP11 binding motifs, bioinformatic analysis of known binding regions can be combined with experimental validation. Although not explicitly described in the search results, the conservation of the THAP DNA-binding domain suggests that THAP11 likely recognizes specific DNA sequence motifs. Comparative analysis of multiple THAP11-bound promoter regions could reveal consensus binding sequences.
Electrophoretic Mobility Shift Assays (EMSA) with purified THAP domain can complement in vivo approaches by defining the direct DNA-binding properties of THAP11. This technique was not explicitly described in the search results but would be valuable for characterizing the sequence specificity and binding affinity of THAP11 to target DNA sequences.
The THAP domain is a putative DNA-binding domain (DBD) that likely also binds a zinc ion. It features a conserved C2CH architecture . The THAP11 protein functions as a transcriptional repressor and plays a crucial role in embryogenesis and the pluripotency of embryonic stem (ES) cells . It acts as a sequence-specific DNA-binding factor that represses gene expression in pluripotent ES cells by directly binding to key genetic loci and recruiting epigenetic modifiers .
THAP11 is involved in various important cellular processes, including epigenetic regulation, maintenance of pluripotency, and transposition . It has been implicated in several disorders, such as methylmalonic aciduria and homocystinuria, Cblj type, and homocystinuria caused by cystathionine beta-synthase deficiency . Additionally, THAP11 has been linked to various cancers .
The human THAP protein family consists of twelve members of different lengths, and THAP11, along with THAP7, has been characterized as a transcription factor . The identification of a predicted oligomerization region in the human THAP protein family opens new directions to investigate the members of this protein family .
In research, recombinant THAP11 protein is used to study its role in transcriptional repression and its involvement in various cellular processes and diseases. Understanding the function and regulation of THAP11 can provide insights into its potential as a therapeutic target for diseases associated with its dysfunction.