Abstract
ABSTRACT
The 26S proteasome is a multi-catalytic protease that serves as the endpoint for protein degradation via the ubiquitin-proteasome system. Proteasome function requires the concerted activity of 33 distinct gene products, but how the expression of proteasome subunits is regulated in mammalian cells remains poorly understood. Leveraging coessentiality data from the DepMap project, here we characterize an essential role for the dystonia gene THAP1 in maintaining the basal expression of PSMB5 . PSMB5 insufficiency resulting from loss of THAP1 leads to defects in proteasome assembly, impaired proteostasis and cell death. Exploiting the fact that the toxicity associated with loss of THAP1 can be rescued upon exogenous expression of PSMB5, we define the transcriptional targets of THAP1 through RNA-seq analysis and perform a deep mutational scan to systematically assess the function of thousands of single amino acid THAP1 variants. Altogether, these data identify THAP1 as a critical regulator of proteasome function and suggest that aberrant proteostasis may contribute to the pathogenesis of THAP1 dystonia.Free full text
PPRID: PPR867630
EMSID: EMS196883bioRxiv preprint, version 1, posted 2024 June 13
https://doi.org/10.1101/2024.06.11.598406
Loss-of-function mutations in the dystonia gene THAP1 impair proteasome function by inhibiting PSMB5 expression
Copyright and license information
Abstract
The 26S proteasome is a multi-catalytic protease that serves as the endpoint for protein degradation via the ubiquitin-proteasome system. Proteasome function requires the concerted activity of 33 distinct gene products, but how the expression of proteasome subunits is regulated in mammalian cells remains poorly understood. Leveraging coessentiality data from the DepMap project, here we characterize an essential role for the dystonia gene THAP1 in maintaining the basal expression of PSMB5. PSMB5 insufficiency resulting from loss of THAP1 leads to defects in proteasome assembly, impaired proteostasis and cell death. Exploiting the fact that the toxicity associated with loss of THAP1 can be rescued upon exogenous expression of PSMB5, we define the transcriptional targets of THAP1 through RNA-seq analysis and perform a deep mutational scan to systematically assess the function of thousands of single amino acid THAP1 variants. Altogether, these data identify THAP1 as a critical regulator of proteasome function and suggest that aberrant proteostasis may contribute to the pathogenesis of THAP1 dystonia.
Introduction
Regulated protein degradation is essential for cellular homeostasis. As the primary route through which the cell achieves selective protein degradation, the ubiquitin-proteasome system (UPS) plays an important role in essentially all critical cellular processes1. Proteins destined for degradation are typically identified by E3 ubiquitin ligases, which, following activation of ubiquitin by an E1 activating enzyme and its transfer to an E2 ubiquitin conjugating enzyme, catalyze the attachment of ubiquitin onto substrate proteins2. Addition of further ubiquitin moieties generates polyubiquitin chains, which can serve as a potent recognition signal for the 26S proteasome, a multi-catalytic protease. The importance of this pathway is underscored by the fact that dysregulation of the UPS is a hallmark of diseases such as cancer, autoimmunity and neurodegeneration3.
The 26S proteasome is a large, multi-subunit complex comprising the 20S core particle and two 19S regulatory particles4. The regulatory subunits are responsible for the recognition and unfolding of ubiquitinated proteins, which are then threaded into the active site in the core particle formed by two rings of β-subunits5. The complex comprises three catalytic subunits: PSMB5 (also known as β5), PSMB6 (β1) and PSMB7 (β2). These exhibit trypsin-like, chymotrypsin-like and caspase-like activities respectively, resulting in the proteolysis of the polypeptide chain into short peptides fragments6. Each catalytic subunit harbors a catalytic threonine residue at its N-terminus6, which is activated following autocatalytic processing of an N-terminal propeptide at a late stage in core particle assembly7.
By performing genome-wide pooled CRISPR/Cas9 loss-of-function genetic screens across hundreds of cancer cell lines, the Broad Institute’s Cancer Dependency Map (DepMap) project aims to systematically catalogue the essentiality of all protein-coding human genes8. A key insight from these data is that whilst the dependency of different cell lines on any one particular gene may vary, genes which function in concert in a biological pathway often exhibit globally similar essentiality patterns9. Thus, by measuring gene dependency across hundreds of cell lines, genes exhibiting ‘co-essential’ relationships can be clustered into modules which may have the power to predict novel functions for genes. Indeed, multiple studies have exploited this dataset to provide new insights into gene function across a range of biological processes9–14.
Here we leveraged insight from co-essentiality data to characterize an essential role for THAP1 in proteasome function. THAP1 is a ubiquitously expressed transcription factor which achieves sequence-specific DNA binding via an atypical THAP-type zinc finger domain located at its N-terminus15. Its target genes remain poorly defined, but THAP1 is thought to play an important role in DNA repair16, cell cycle progression17 and oligodendrocyte myelination18,19. Homozygous deletion of THAP1 leads to embryonic lethality 20,21. Notably, a wide variety of autosomal dominant mutations located throughout the THAP1 coding sequence cause an early-onset form of the neurological disorder dystonia (DYT-THAP1, previously known as DYT-6), where progressive loss of motor function leads to sustained involuntary muscle contractions and abnormal posturing22,23. However, as the critical targets of THAP1 are poorly characterized, it remains unclear how the THAP1 mutations observed in dystonia patients result in disease.
Here, by interrogating the co-essential relationship between THAP1 and PSMB5, we characterize an essential role for THAP1 in proteasome function. Exploiting a fluorescent reporter knocked into the endogenous PSMB5 locus, we demonstrate that the co-essential relationship between THAP1 and PSMB5 is explained by an essential role for THAP1 in activating PSMB5 expression. THAP1 binds to cognate sites within the PSMB5 promoter and is required for its basal expression, and hence loss of THAP1 results in insufficient PSMB5 expression, proteasome dysfunction and cell death. Finally, we leveraged our functional reporter assay to perform a deep mutational scan of THAP1, quantifying the activity of thousands of single amino acid variants to define the landscape of THAP1 mutations in dystonia.
Results
The dystonia gene THAP1 exhibits a co-essential relationship with the proteasome subunits PSMB5 and PSMB6
Leveraging co-essentially data from the DepMap project8, we set out to characterize novel roles for genes involved in the UPS. Focusing on a manually curated set of ~1000 genes implicated in UPS function, we examined co-essential gene relationships derived from genome-wide CRISPR-Cas9 screens across ~1100 different cancer cell lines. Supporting the utility of this approach to identify genetic relationships that are functionally relevant, many of the most significant positive co-essential relationships clustered genes whose products are known to act in multi-protein complexes to facilitate protein degradation (Fig. S1A). For instance, the RNF126 E3 ubiquitin ligase cooperates with BAG6 to target hydrophobic sequences mislocalised to the cytosol for proteasomal degradation24 (Fig. 1A); Cul2, ElonginB/C and the von Hippel-Lindau (VHL) substrate adaptor comprise a Cullin-RING E3 ubiquitin ligase complex responsible for the degradation of hypoxia-inducible factor (HIF)-1α in normoxia (Fig. 1B)25, and the CTLH complex is a multi-subunit E3 ligase orthologous to the yeast GID complex which degrades gluconeogenic enzymes26 (Fig. 1C). Furthermore, several of the most significant negative co-essential relationships define E3 ligase-substrate pairs: for example, MDM2 mediates the degradation of p5327 (Fig. S1B) and the Cul4 substrate adaptor AMBRA1 targets cyclin D28–30 (Fig. S1C).
Our follow-up work focused on the most statistically significant uncharacterized co-essential relationship in the dataset: THAP1 exhibits a highly significant positive association with both PSMB5 and PSMB6 (Fig. 1D-F). THAP1 is a transcription factor which binds DNA in a sequence-specific manner using a THAP-type zinc-finger domain, while PSMB5 and PSMB6 encode catalytic subunits of the proteasome core particle. Thus, we set out to test the hypothesis that the co-essential relationship between THAP1 and PSMB5/6 could be explained by an essential role for THAP1 in regulating the expression of catalytic proteasome subunits.
Loss of THAP1 abrogates PSMB5 transcription
Lentiviral expression of Cas9 and CRISPR sgRNAs targeting THAP1 in HEK-293T cells was extremely toxic (Fig. 1G-H), consistent with DepMap data which demonstrates that knockout of THAP1 is broadly deleterious across cancer cell lines8. However, at day 5 post-transduction, before the onset of significant cell death, we found substantially reduced levels of PSMB5 transcripts by quantitative reverse transcription PCR (qRT-PCR) (Fig. 1I). In contrast, we observed no reduction in the expression of either PSMB6 or PSMB7, the other catalytic subunits of the proteasome (Fig. 1I). Concordantly, we also observed reduced abundance of PSMB5 protein as assessed by immunoblot (Fig. 1J). Thus, these data suggest that THAP1 is required to maintain basal levels of PSMB5 transcription.
Lethality resulting from loss of THAP1 can be rescued by exogenous PSMB5
Next, we sought to test the hypothesis that the essentiality of THAP1 is due to its role in activating PSMB5 expression. Should this be the case, we reasoned that, irrespective of any reduction in the expression of endogenous PSMB5, an exogenous source of PSMB5 should rescue cell viability upon THAP1 ablation. Strikingly, unlike their wild-type counterparts, HEK-239T cells transduced with a lentiviral vector expressing PSMB5 did not display any significant growth defect following CRISPR/Cas9-mediated targeting of THAP1 (Fig. 2A and Fig. S2A). Exogenous expression of PSMB6, however, was incapable of rescuing viability following THAP1 ablation (Fig. 2A and Fig. S2B). Thus, the toxicity that results from loss of THAP1 is due to insufficient PSMB5 expression, explaining the molecular basis for their co-essential relationship. In contrast, we found no evidence to support a direct relationship between THAP1 and PSMB6, suggesting that their co-essential relationship arises indirectly through their shared relationship with PSMB5.
DepMap data demonstrates that disruption of THAP1 is broadly lethal across cancer cell lines (Fig. 2B). Thus, to generalize our finding that the essential requirement for THAP1 is to facilitate PSMB5 expression, we ablated THAP1 in three additional human cell lines: HeLa, A549 and THP-1. Mirroring our findings in HEK-293T cells, in A549 and HeLa we found that the toxicity observed upon loss of THAP1 could be ameliorated upon exogenous expression of PSMB5 (Fig. 2C-D). THP-1 cells, however, did not exhibit reduced viability following THAP1 disruption (Fig. 2E). This prompted us to examine in more detail the nature of the cell lines in which THAP1 is not essential, which were strikingly enriched (P < 1x10-9) for immune cells (‘myeloid’ or ‘lymphoid’ as defined by DepMap). Considering that the immunoproteasome is constitutively expressed by many immune cells31,32, we reasoned that expression of PSMB8, the analogous counterpart of PSMB5 in the immunoproteasome, might relieve the essential requirement for THAP1. Indeed, there is a strong correlation between the essentiality of THAP1 and PSMB8 expression levels as measured by RNA-seq, wherein the cell lines in which THAP1 knockout has little or no impact on viability express the highest levels of PSMB8 (Fig. 2F). Indeed, we found that THP-1 cells expressed high levels of PSMB8 by qRT-PCR (Fig. 2G). We further validated these conclusions in HEK-293T cells, where, like PSMB5, exogenous expression of PSMB8 maintained the viability of THAP1 knockout cells (Fig. 2H and Fig. S2C). Thus, sustained expression of either PSMB5 or its immunoproteasome counterpart PSMB8 can rescue the toxicity associated with loss of THAP1.
A fluorescent reporter at the endogenous PSMB5 locus monitors THAP1 activity in live cells
We extended these findings by knocking-in a fluorescent reporter to the endogenous PSMB5 locus, enabling us to monitor PSMB5 expression in live cells. Following transfection of HEK-293T cells with Cas9, an sgRNA targeting the transcriptional start site of PSMB5 and a homology donor vector encoding the green fluorescent protein (GFP) variant mClover333 followed by a 2A peptide (Fig. 3A), we were readily able to establish a population of cells (~10%) which were stably GFP-positive (Fig. 3B). Single cell clones isolated from the GFP-positive population (Fig. 3C) harbored GFP at the intended site as validated by PCR from genomic DNA (Fig. S2D). Furthermore, lentiviral expression of shRNAs targeting PSMB5 resulted in a reduction in GFP expression (Fig. S2E), validating that the reporter clones could be used to quantitatively assess PSMB5 expression.
We exploited our findings above to generate viable THAP1 knockout (KO) reporter cells. Following CRISPR/Cas9-mediated disruption of THAP1 in PPSMB5-GFP reporter cells (sustained by exogenous expression of PSMB5) (Fig. 3D), we isolated single cell clones from the GFPdim population by fluorescence-activated cell sorting (FACS) (Fig. 3E). Disruption of the THAP1 locus was confirmed by PCR from genomic DNA followed by Sanger sequencing (Fig. S2F). Using primers specific to the 3’ untranslated region of PSMB5 to allow for selective detection of the endogenous transcript, we confirmed a substantial reduction in PSMB5 expression in two THAP1 KO clones by qRT-PCR (Fig. 3F). However, our attempts to validate efficient knockout of THAP1 by immunoblot were hampered by the paucity of effective commercial antibodies. In particular, we found that the Proteintech antibody (12584-1-AP) used in multiple previous studies detected a prominent band running around the expected molecular weight (~25 kDa), but whose abundance was not affected upon CRISPR/Cas9 targeting of THAP1 (Fig. 3G). However, this antibody could readily detect exogenous THAP1 as a separate band that migrated just slightly slower, and, upon prolonged exposure, was able to detect endogenous THAP1 in control cells but not in the THAP1 knockout clones (Fig. 3G).
THAP1 acts through cognate binding sites located within the PSMB5 promoter
Next, we sought to determine how THAP1 might regulate PSMB5 expression. In support of a direct effect, chromatin immunoprecipitation followed by sequencing (ChIP-seq) data from the ENCODE project34 revealed THAP1 occupancy immediately upstream of the PSMB5 transcription start site (TSS) (Fig. 4A). Despite THAP1 binding to thousands of gene promoters (Table S1), this property is not shared among the genes encoding proteasome subunits: PSMD8 is the only other gene to exhibit THAP1 occupancy (Fig. 4A). THAP1 contains a THAP-type zinc finger domain which mediates sequence-specific DNA binding, and competitive EMSA experiments35 have defined the consensus binding sequence (“THABS” motif) as TNNNGGCA (where N represents any nucleotide) (Fig. 4B). Strikingly, examination of the PSMB5 proximal promoter region revealed two perfect matches within 200 bp of the TSS, and a third near-perfect match a further ~500 bp upstream (Fig. 4C). Together, these data suggest that THAP1 binds cognate motifs in the PSMB5 promoter to activate its transcription.
We examined this hypothesis by engineering a lentiviral reporter system in which ~1 kb of the PSMB5 proximal promoter was placed upstream of GFP (Fig. 4D). Single copy expression of this reporter construct in HEK-293T cells resulted in robust GFP expression (Fig. S3A). This appeared to be due in part to the activity of THAP1, as combined deletion of all three THABS motifs from the PSMB5 promoter (“ΔTHABS”) resulted in decreased GFP expression (Fig. 4E). Importantly, CRISPR-mediated ablation of THAP1 (performed following the introduction of exogenous PSMB5 to maintain cell viability) reduced GFP expression from the reporter construct driven by the wild-type PSMB5 promoter, but did not further reduce expression from the PSMB5 promoter lacking all THABS sites (Fig. 4F).
To investigate the relative importance of the three THABS motifs, we created an additional panel of mutant constructs in which each THABS motif was individually deleted. These data pointed to a critical role for site 2, as only the ΔTHABS2 construct exhibited decreased GFP expression relative to the level of the ΔTHABS construct (Fig. S3B). Moreover, the ΔTHABS2 construct was the only one unaffected following ablation of THAP1, whereas a marked reduction in GFP expression was observed in cells expressing ΔTHABS1 and ΔTHABS3 (Fig. S3C). Thus, THAP1 binding to a cognate motif (THABS2) immediately upstream of the PSMB5 TSS appears critical for PSMB5 expression.
Loss of THAP1 impairs proteasome function
As PSMB5 encodes one of the three catalytic subunits of the constitutive 20S proteasome core particle, we set out to test the hypothesis that the toxicity associated with loss of THAP1 was due to proteasome dysfunction. First, we examined whether the catalytic activity of PSMB5 was required to sustain cell viability upon THAP1 ablation. Whereas HEK-293T cells expressing wild-type PSMB5 did not exhibit any appreciable growth defect upon disruption of THAP1, cells expressing a catalytically-inactive PSMB5 mutant were not viable under these conditions (Fig. 5A). Second, as PSMB5 is critical to facilitate the integration of the other catalytic β subunits into the 20S core particle during proteasome assembly36–38, we assessed whether loss of THAP1 resulted in defects in proteasome assembly. As a result of impaired autocatalytic cleavage of their N-terminal propeptide, an inability of the catalytic subunits to incorporate into the 20S core particle causes an accumulation of the immature proteins which can be detected by immunoblot38. Supporting the idea of defective proteasome assembly in the absence of THAP1, we found decreased abundance of the mature forms of PSMB6 and PSMB7, concomitant with the accumulation of immature (uncleaved) species (indicated by asterisks) which were absent from control cells (Fig. 5B). Finally, we reasoned that proteasome dysfunction upon THAP1 loss should result in the stabilization of short-lived proteins. Exploiting the Global Protein Stability (GPS) two-color lentiviral reporter system39 (Fig. 5C), we found that CRISPR-mediated ablation of THAP1 stabilized two exogenous GFP-degron fusion proteins to a similar degree as shRNA-mediated knockdown of PSMB5 (Fig. 5D). Similarly, THAP1 disruption also resulted in increased abundance of endogenous HIF-1α, which is constitutively degraded by the proteasome in normoxia25,40 (Fig. 5E). Altogether, these data support a model whereby the death of cells lacking THAP1 is caused by defective proteasome function resulting from inadequate expression of PSMB5.
Defining the transcriptional targets of THAP1
The key transcriptional targets of THAP1 remain poorly defined, hampering our ability to understand the functional consequences of THAP1 mutations in disease. Armed with the knowledge that exogenous PSMB5 expression ameliorates the toxicity associated with THAP1 loss, we reasoned that we could leverage our findings to assess the impact of THAP1 deletion on the transcriptome. To avoid potential artefacts resulting from the analysis of single cell clones, we purified a population of THAP1 knockout cells. Following the CRISPR-mediated ablation of THAP1 in PSMB5-GFP knock-in reporter cells (overexpressing PSMB5 to ensure viability), we isolated GFPdim cells by FACS and performed RNA-seq analysis (Fig. 6A). After discounting genes exhibiting differential expression between untransduced cells and cells expressing control sgRNAs, we identified 277 genes (220 downregulated, 57 upregulated) whose expression was significantly altered (FDR < 0.001, fold-change > 2) upon THAP1 knockout (Fig. 6B and Table S2). Supporting the veracity of the dataset, the most significantly downregulated gene was Shieldin complex subunit 1 (SHLD1, previously known as C20orf196), consistent with recent findings describing a role for THAP1 in DNA double strand break repair choice16. Furthermore, although the requirement for exogenous expression of PSMB5 precluded its identification as a differentially expressed gene, the abundance of intronic reads mapping to the endogenous PSMB5 locus was greatly reduced in the THAP1 knockout cells (Fig. S4A-B).
To identify direct transcriptional targets of THAP1, we cross-referenced the differentially expressed genes identified through RNA-seq with THAP1 binding sites as defined by ChIP-seq. Among the differentially expressed genes, 42 exhibited THAP1 occupancy in their proximal promoter (Fig. 6C); of these 42 direct targets, 19 were downregulated upon loss of THAP1 while 23 were upregulated, suggesting that THAP1 has the potential to act as either a repressor or activator of transcription depending on the genomic context. However, the primary role of THAP1 appeared to be as an activator, with several of its direct targets exhibiting marked downregulation in its absence (Fig. 6B-C). We found no significant functional enrichment among the differentially expressed genes through GO term analysis, but their promoter sequences were enriched for transcription factor binding motifs for both THAP1 and YY1, a known THAP1 co-factor16,41,42 (Fig. 6D and Fig. S4C). None of these genes are currently associated with dystonia, but they include ECH1, an enzyme involved in fatty acid metabolism that has been previously identified as a THAP1 target42, and METTL3, the N6-methyladenosine methyltransferase, which is an attractive therapeutic target in cancer43. Across five of the THAP1 target genes that exhibited the greatest degree of downregulation upon loss of THAP1, we further validated these findings by qRT-PCR (Fig. 6E-I). Altogether, these data define the genes directly targeted by the transcription factor activity of THAP1.
A deep mutagenic scan defines the landscape of THAP1 mutations in Dystonia
A wide range of autosomal dominant mutations distributed throughout the THAP1 coding sequence give rise to DYT-THAP1 dystonia44–50, an early-onset neurological disorder characterized by involuntary muscle contractions and movements causing abnormal and painful posturing. Thus, we sought to exploit our PPSMB5-GFP knock-in reporter clone to assess the functional impact of DYT-THAP1 mutations. Genetic complementation of THAP1 KO cells with wild-type THAP1 did result in a restoration in PPSMB5-GFP expression, although this effect was partial and did not restore GFP fluorescence to the levels observed in the parental cells (Fig. S5A). However, this assay was sufficiently sensitive to report on THAP1 activity, as expression of an inactive THAP1 mutant unable to bind DNA (C5A, which abrogates zinc chelation by the zinc finger motif15) did not restore PPSMB5-GFP expression (Fig. S5A).
With the goal of globally defining how mutations in THAP1 affect its function, we leveraged our phenotypic assay in PPSMB5-GFP knock-in reporter cells to carry out a deep mutational scan. Through microarray oligonucleotide synthesis, we generated a library of mutant constructs in which each residue of THAP1 (with the exception of the initiator methionine, 212 amino acids total) was systematically replaced with all of the other 20 possible amino acids (Fig. 7A-B). The resulting site-saturation mutagenesis library was packaged into lentiviral particles and introduced into THAP1 KO PPSMB5-GFP reporter cells at low multiplicity of infection, ensuring single-copy expression. We then used FACS to partition the population into GFPdim cells, in which no restoration of PPSMB5-GFP expression was observed, and GFPbright cells, in which PPSMB5-GFP expression was restored, and quantified the THAP1 variants present in each population by Illumina sequencing (Fig. 7B).
After an initial filtering step to remove variants with low read counts, we recovered data for 4002 of the 4240 possible single amino acid variants (94.3%). Overall, we observed high concordance between mutant performance across two replicate experiments (Fig. S5B); however, we discarded 179 mutants which exhibited discordant behavior between the two replicate experiments, leaving a total of 3823 (90.2%) variants for analysis (Table S3). The results are summarized as a heatmap in Fig. 7C, with the data normalized such that the mean performance of all the control (wild-type) constructs is centered at 1; thus, the darker the red color the more deleterious the impact of the mutation on THAP1 function, whereas blue cells indicate mutations which may enhance the THAP1-mediated activation of PPSMB5-GFP expression.
We evaluated the quality of the dataset in several ways. First, we considered residues essential for zinc chelation and hence folding of the zinc finger motif15: C5, C10, C54 and H57. These critical residues were uniformly essential for THAP1 activity, as mutation to any other residue prevented activation of the PPSMB5-GFP reporter (Fig. 8A). Moreover, mutations across all residues previously determined to be important for DNA binding through biochemical assays15 were extremely deleterious (Fig. 8B). Second, the global landscape of THAP1 activity correlated well with the predicted structure of THAP1 (Fig. 8C-D). In particular, mutation of most residues in the two structured regions, the THAP-type zinc finger (residues 2-81) and the predicted coiled-coil domain (residues 139-191) abrogated transcriptional activity, whereas most mutations targeting the unstructured central linker (residues 82-138) and C-terminus (residues 192-213) did not impair transcriptional activity (Fig. 7C and Fig. 8E). A notable exception, however, was the DHNY motif (residues 134-137) lying at the end of the central unstructured linker, which was absolutely critical for THAP1 function (Fig. 7C). Interestingly, this motif has been identified as the binding site for HCFC151, an essential cofactor for the THAP1-mediated activation of SHLD16. Thus, the interaction with HCFC1 is also likely to be critical for the THAP1-mediated activation of PSMB5. Indeed, we confirmed that deletion of the DHNY motif and the coiled-coil domain, but not the disordered C-terminus, abolished the ability of THAP1 to activate the PPSMB5-GFP reporter (Fig. 8F).
Many of the THAP1 mutations identified in dystonia patients remain of uncertain significance50. Thus, we examined the utility of this dataset in classifying the functional effects of THAP1 variants identified clinically (Fig. 8G). The majority of these mutations strongly impaired the ability of THAP1 to activate expression of the PPSMB5-GFP reporter, consistent with the prevailing view that disease-causing mutations represent loss-of-function alleles19,41,50. However, some mutants exhibited activity at or approaching the level of the wild-type protein, suggesting that they might represent benign variants. To verify that the screen results could be faithfully recapitulated in individual experiments, we selected eight patient mutations predicted to abolish THAP1 activity (A7D, R13H, K24E, P26R, H57N, L72R, F81L and N136S) and compared their performance to five mutants predicted not to affect THAP1 activity (I80V, C83R, M143V, A166T and D192N). Validating the screen results, the eight inactive mutants exhibited little or no ability to activate PPSMB5-GFP reporter expression (Fig. 8H and Fig. S5C-E), whereas the five active mutants exhibited similar performance to wild-type THAP1 (Fig. 8H). Altogether, these data illustrate structure-function relationships for THAP1 at high resolution, enabling the functional classification of clinical THAP1 mutations.
Discussion
Co-essential relationships identified through the DepMap project8 represent a rich resource to characterize gene function. Here we explain the co-essential relationship between THAP1 and PSMB5 by demonstrating that THAP1 is essential for the basal expression of PSMB5. Insufficient PSMB5 expression resulting from loss of THAP1 results in proteasome dysfunction and cell death, which can be rescued through exogenous expression of PSMB5. We exploit this finding to generate viable THAP1 knockout cells and hence identify transcriptional targets of THAP1 by RNA-seq. Finally, leveraging a phenotypic assay to systematically assess the activity of THAP1 mutants at the endogenous PSMB5 locus, we define the transcriptional activity of THAP1 mutants found in dystonia patients. Overall, these data identify THAP1 as a novel regulator of proteasome function and suggest that aberrant proteostasis could be a factor underlying the pathogenesis of THAP1 dystonias.
Two major players are known to regulate the expression of proteasome subunits. In yeast the transcription factor Rpn4 is responsible for both the basal expression of proteasome subunits, and, under conditions of proteasome insufficiency, feedback induction of proteasome subunit expression52. Rpn4 functions as part of a negative feedback loop that monitors proteasome activity: in unstressed cells Rpn4 is constitutively degraded, but it is rapidly stabilized upon proteasome dysfunction53. In mammalian cells Nrf1 acts in a similar manner to induce the expression of proteasome subunits under conditions of proteasome insufficiency54,55, but appears not to have a major role in their basal expression54. Several other transcription factors have been implicated in proteasome gene expression56–58, including NF-Y which regulates a set of proteasome genes which carry a CCAAT box motif in their promoters58, but the factors which maintain the basal expression of proteasome subunits in human cells remain largely unknown.
Here we characterize THAP1 as an additional regulator of proteasome gene expression. In contrast to the master regulators Rpn4 and Nrf1, THAP1 appears to regulate only PSMB5. Why might THAP1 have evolved to exclusively regulate the expression of one single proteasome subunit? We speculate that perhaps there is a physiological circumstance wherein the downregulation of PSMB5 expression is beneficial, which could be achieved through the conditional inactivation of THAP1 activity. For example, it is plausible that transcriptional downregulation of PSMB5 concomitant with upregulation of PSMB8 might be beneficial upon viral infection, when immunoproteasomes are favored to increase the production of antigenic peptides59,60, or during thymic development when PSMB11 (β5t) is incorporated in preference to PSMB5 and PSMB8 into the thymoproteasome61. Another interesting question for future studies will be to examine whether the expression of other individual proteasome subunits (such as PSMB6 and PSMB7) is also subject to specific regulatory mechanisms.
The mechanisms through which THAP1 mediates its effects on gene expression remain unclear. THAP1 does not possess an obvious activator or repressor domain, and so it is likely that it acts through the recruitment of co-factors to target genes. A complex of THAP1 with YY1 and HCFC1 has previously been shown to mediate activation at the SHLD promoter16, and the F81L dystonia mutation is thought to disrupt YY1 binding and hence impair THAP1-mediated transcriptional activation42. Our data offer support to this notion: YY1 binding motifs were strongly enriched amongst the direct targets of THAP1 identified by RNA-seq, and an intact HCFC1-binding motif was critical for THAP1-mediated activation of the PPSMB5-GFP reporter. However, ChIP-seq reveals thousands of THAP1 binding sites in promoters across the genome, many of which colocalize with HCFC1 and YY116, and yet we observed relatively few transcriptional changes by RNA-seq. Thus, we speculate that other factors must be involved in determining whether THAP1 binding alters transcriptional activity. Our PPSMB5-GFP knock-in reporter cells may therefore serve as a useful resource for further genetic interrogation of this pathway; for example, genome-wide CRISPR screens may identify additional genes required for the THAP1-mediated activation of PSMB5.
We leveraged our genetic reporter to characterize the impact of single amino acid variants on the ability of THAP1 to activate the expression of PSMB5. This dataset, covering over 90% of all possible single amino acid variants, represents a rich resource for functional classification of THAP1 mutations. Specifically, these data strongly support the notion that disease-causing mutations in THAP1 generate loss-of-function alleles which are unable to regulate target gene expression: 84% of the missense mutations in THAP1 identified in dystonia patients exhibited performance at <50% of the wild-type protein. Thus, we propose that the mutations which do not impair THAP1 activity are likely to represent benign variants. Indeed, the clinical evidence supporting a pathogenic role for some THAP1 mutations remains equivocal50; for example, a previous study concluded that the I80V mutation was very likely to be benign, supported by the conservative nature of the substitution and the lack of any functional defect in a reporter assay50.
How does loss of THAP1 function result in Dystonia? As the most plausible explanation is that dysregulated expression of one or more of its target genes leads to disease62, our data advance progress towards answering this question in two ways. First, the identification of THAP1 as a critical activator of PSMB5 expression suggests that proteasome dysfunction could underlie the pathogenesis of DYT-THAP1. Second, by exploiting exogenous PSMB5 expression to generate viable THAP1 knockout cells, we were able to rigorously identify additional direct transcriptional targets of THAP1. However, as none of these genes are currently associated with dystonia and aberrant proteostasis is a feature of many neurological disorders63,64, these data highlight proteasome dysfunction as a candidate pathogenic mechanism underlying THAP1 dystonias.
Materials & Methods
Cell culture
HEK-293T, HeLa and A549 cells were grown in Dulbecco’s Modified Eagle’s Medium (DMEM, Merck #D6429); Jurkat and THP-1 cells were grown in Roswell Park Memorial Institute Medium (RPMI, Merck #R8758). Both were supplemented with 10% fetal bovine serum (ThermoFisher Scientific, #A5256701) plus penicillin and streptomycin (ThermoFisher Scientific, #15140122) and incubated at 37°C plus 5% CO2. All cells were routinely checked for mycoplasma contamination.
Antibodies
Primary antibodies used in this study were: rabbit anti-THAP1 (Proteintech, #12584-1-AP), mouse anti-V5 tag (Abcam, #AB27671), rabbit anti-PSMB5 (Enzo Life Sciences, #BML-PW8895), mouse anti-PSMB6 (Enzo Life Sciences, #BML-PW8140), mouse anti-PSMB7 (Enzo Life Sciences, #BML-PW8145), mouse anti-HIF-1α (BD, #610959), mouse anti-Vinculin (Sigma, #V9131) and mouse anti-β-actin (Sigma, #A2228). HRP-conjugated donkey anti-mouse IgG and donkey anti-rabbit IgG secondary antibodies were obtained from Jackson ImmunoResearch.
Plasmids
Proteasome 20S core particle β-subunits were exogenously expressed from the pHRSIN-PSFFV-GFP-WPRE-PPGK-BlastR/HygroR lentiviral vectors (a gift from Paul Lehner), with constructs cloned in place of GFP via the Gibson assembly method using the NEBuilder HiFi Cloning Kit (NEB, #E5520S). GPS lentiviral vectors encoding the N-terminus of PTGS1 and the C-terminus of TNNC2 fused to GFP were gifts from Stephen Elledge. CRISPR sgRNA sequences were selected from the Brunello genome-wide library65 and synthesized as top and bottom strand oligonucleotides (IDT). Oligos were phosphorylated (T4 PNK; NEB #M0201), annealed by heating to 95°C followed by slow cooling to room temperature, and then inserted (T4 ligase; NEB #M0202) into lentiCRISPRv2 (Addgene #52961). shRNAs were cloned in an analogous manner into the pHR-SIREN-PU6-shRNA-WPRE-PPGK-Puro lentiviral vector (a gift from Paul Lehner) using the BamHI and EcoRI sites. Top strand oligonucleotide sequences used were:
sg1-Control (targets FOXP1 intron): caccgTGGGAACAGGATGAGGAAGG
sg2-Control (targets ATP1A1 intron): caccGATGGGCAAGAAGGAAGCAG
sg1-THAP1: caccgCTGCAAGAACCGCTACGACA
sg2-THAP1: caccGAAAACTGAGAGATTAACAG
sg3-THAP1: caccgCTGTGACCACAACTATACTG
shControl:gattcGTTATAGGCTCGCAAAAGGTTCAAGAGACCTTTTGCGAGCC TATAACTTTTTTg
shPSMB5:gattcCAATGTCGAATCTATGAGCTTCTCGAGAAGCTCATAGATTC GACATTGTTTTTTg
Lentivirus production
Lentiviral stocks were generated through the transfection of HEK-293T cells with the specific lentiviral vector plus a mix of packaging plasmids encoding Gag-Pol, Rev, Tat and VSV-G. HEK-293T cells seeded at 70-90% confluence were transfected using PolyJet In Vitro DNA Transfection Reagent (SignaGen Laboratories, #SL100688) according to the manufacturer protocol. The media was replaced 24 hours post-transfection and the viral supernatant was collected at 48 hours post-transfection, centrifuged at 800 x g for 5 minutes to remove cellular debris, and either applied immediately to target cells or stored at -80°C in single-use aliquots.
CRISPR/Cas9-mediated gene knock-in
A four-fragment Gibson assembly reaction was used to generate the homology donor vector. 5’ and 3’ homology arms (~1 kb) were amplified from genomic DNA, and were assembled together with a fragment encoding mClover3 followed by a 2A peptide and a pUC plasmid digested with PciI (NEB, #R0655) and SbfI (NEB, #R3642). The resulting plasmid was transfected into HEK-239T cells along with a PX459 (Addgene #48139, kindly deposited by Feng Zhang) plasmid encoding Cas9 and an sgRNA (CTTTCTGCCCACACTAGACA) targeting the start of the PSMB5 coding sequence. Transfected cells were selected with puromycin for 48 h commencing 24 h post-transfection. Two weeks later, cells that remained GFP+ were single cell cloned by FACS.
Flow cytometry and FACS
Analysis of cells by flow cytometry was performed using either an LSR-II or Fortessa instrument (BD Biosciences), collecting a minimum of 10,000 cells per sample. All flow cytometry data were collected through FACSDiva software and subsequently analyzed using FlowJo. Cell sorting was carried out using an Influx instrument (BD Biosciences).
Immunoblotting
Cells were lysed in 1% SDS plus 1:200 Benzonase (Merck, #E1014) for 20 minutes at room temperature. Following the addition of Laemmli buffer (Bio-Rad, #161-0747), lysates were heated to 70°C for 10 minutes. Proteins were separated by SDS-PAGE using 4-12% Bis-Tris gels (Merck, #MP41G12) and transferred onto an activated PVDF membrane (Merck, #IPFL00010). Membranes were blocked for a minimum of 30 min in 5% Skim Milk Powder (Merck, #70166) in PBS + 0.1% Tween-20 (PBS-T) (Merck, #P1379). Membranes were incubated with primary antibodies overnight at 4°C, washed at least three times in PBS-T, and then incubated with HRP-conjugated secondary antibodies for 40 minutes at room temperature. Following a further five washes in PBS-T, reactive bands were visualized using SuperSignal West Detection Reagents (ThermoFisher Scientific, #32106, #34580 and #34076) and images collected on a ChemiDoc Imaging System (Bio-Rad). Raw images were processed using GNU Image Manipulation Platform (GIMP) version 2.10.34.
Imaging
HEK-293T cells were imaged on a Zeiss Primovert Inverted Phase Contrast Microscope Ph1/0.3 at 10x magnification using the NexYZ 3-axis Universal Smartphone Adapter (Celestron).
qRT-PCR
Total RNA was extracted from ~1 million cells using the RNeasy Mini Kit (Qiagen, #74104) with QIAshdredder Mini Spin Columns (Qiagen, #79656) as per the manufacturer’s protocol, including on-column DNaseI digestion using the RNase-Free DNase Set (Qiagen, #79254). Reverse transcription was performed with 1 μg of RNA using one-step reaction using LunaScript RT SuperMix Kit (NEB, #E3010) as indicated by the manufacturer. For subsequent analysis by qPCR, 1 μl of cDNA template, 0.5 μl of each primer (10 μM) and 12.5 μl Luna Universal Probe qPCR Master Mix (NEB, M3004) were mixed in a final volume of 25 μl; thermocycling was performed on a QuantStudio 7 Flex Real-Time PCR system (ThermoFisher Scientific). Relative expression was quantified using the ΔΔCt method relative to RPS18; data are expressed as mean ± standard deviation and P values calculated using a one-tailed unpaired t-test. All qPCR amplicons were verified using agarose gel electrophoresis. Primer sequences are listed in Table S4.
RNA-seq
RNA extracted as above was sent to Azenta for strand-specific polyA+ Illumina library preparation and sequencing. Raw sequence reads were trimmed of adaptor sequence using Cutadapt (version 4.1), aligned using HISAT2 (version 2.2.1) to the human genome (GRCh38 genome_tran index), and further analyzed using SeqMonk (version 1.48.1).
Deep mutational scan
The THAP1 coding sequence was divided into six segments for mutagenesis (encompassing residues 2-37, 38-73, 74-109, 110-145, 146-181 and 182-213). Using pKLV2-PEF1α-THAP1-PPGK-Puro-2A-BFP-WPRE as the starting point, six vectors were generated in which ‘stuffer’ regions flanked by BbsI restriction sites replaced the sequence encoding each segment. An oligonucleotide pool encoding the mutant alleles was synthesized by Twist Bioscience: for each segment, each amino acid was systematically exchanged to all 20 possible amino acids. Each of the six mutant segments were amplified from the oligonucleotide pool by PCR (Q5, NEB #M0491L), gel purified (QIAEX II Gel Extraction Kit, Qiagen #20021), and then cloned into their respective ‘stuffer’ vector cut with BbsI (NEB, #R3539S) using the Gibson assembly method (NEB, #E5520S). The reaction products were electroporated into DH10β cells (ThermoFisher Scientific, #18290015) and grown on LB plates with ampicillin overnight at 30°C; the next morning, plasmid DNA was extracted (GenElute HP Plasmid Midiprep Kit, Merck #NA0200-1KT) from all of the E. coli and verified by Sanger sequencing (Azenta).
The six mutant pools were combined into three for screening (1-2, 3-4 and 5-6). These were packaged into lentiviral particles and, in duplicate, introduced into GFPdim THAP1 KO PPSMB5-GFP reporter cells at a multiplicity of infection of ~0.3 (~30% BFP+ cells). Five days post-transduction, the BFP+ cells were partitioned into GFPdim (THAP1 inactive) and GFPbright (THAP1 active) populations by FACS. Genomic DNA was extracted from the sorted cells (Gentra Puregene Cell Kit, Qiagen #158767) and the exogenous THAP1 sequences in each sample amplified by PCR (Q5, NEB #M0493L), using primers annealing to invariant regions flanking each mutagenized segment. PCR products were purified using a spin column (QIAquick PCR Purification Kit, Qiagen #28104), and then used as a template for a second PCR reaction using primers to add the Illumina adaptors and indexes. Products were purified using a spin column, quantified using a Nanodrop spectrophotometer, and mixed evenly; the final pool was purified from a 2% agarose gel (QIAEX II Gel Extraction Kit, Qiagen #20021). All steps were performed at sufficient scale so as to maintain at least 200-fold representation of the library. Sequencing was performed on an Illumina NovaSeq 6000 instrument using 150 bp paired-end reads. Count tables quantifying the abundance of each mutant in each sorting bin were generated by trimming the raw sequence reads of constant flanking sequence using Cutadapt (version 4.1) and aligning them to a reference index using Bowtie 2 (version 2.4.5).
Supplementary Material
- Table S1 Download source data
- Table S2 Download source data
- Table S3 Download source data
- Table S4 Download source data
- Supplementary Information Download source data
Acknowledgements
We are grateful to Gabriela Grondys-Kotarba and Reiner Schulte at the Cambridge Institute for Medical Research Flow Cytometry Core Facility and to the NIHR Cambridge BRC Cell Phenotyping Hub. This work was supported by an Academy of Medical Sciences Springboard Grant (SBF007\100019) and an Isaac Newton Trust/Wellcome Trust ISSF/University of Cambridge Joint Research Grant to R.T.T. R.T.T. is a Pemberton-Trinity Fellow.
Author Information
Correspondence: ku.ca.mac@02ttr
Data Availability
THAP1 RNA-seq data has been deposited at GEO (GSE264536) and Illumina sequencing data from the THAP1 deep mutagenic scan are available at SRA (PRJNA1102672). THAP1 ChIP-seq data was obtained from GEO (GSM803408). DepMap datasets are publicly available at https://depmap.org/portal/download/all/. Additional data and/or reagents that support the findings of this study are available from the corresponding author upon reasonable request.
References
- 1.Ubiquitylation at the crossroads of development and disease. Nature Reviews Molecular Cell Biology. 2017; 19: 59-70. 2017 19:1 [Europe PMC Abstract]
- 2.The Ubiquitin Code in the Ubiquitin-Proteasome System and Autophagy. Trends Biochem Sci. 2017; 42: 873-886. [Europe PMC Abstract]
- 3.Ubiquitination in disease pathogenesis and treatment. Nat Med. 2014; 20: 1242-1253. [Europe PMC Abstract]
- 4.Molecular mechanisms of proteasome assembly. Nature Reviews Molecular Cell Biology. 2009; 10: 104-115. 2009 10:2 [Europe PMC Abstract]
- 5.The Logic of the 26S Proteasome. Cell. 2017; 169: 792-806. https://doi.org/10.1016/j.cell.2017.04.023 [Europe PMC Full Text] [Europe PMC Abstract]
- 6.The proteasome: Overview of structure and functions. Proc Jpn Acad Ser B Phys Biol Sci. 2009; 85: 12. https://doi.org/10.2183/pjab.85.12 [Europe PMC Full Text] [Europe PMC Abstract]
- 7.Autocatalytic subunit processing couples active site formation in the 20S proteasome to completion of assembly. Cell. 1996; 86: 961-972. [Europe PMC Abstract]
- 8.Defining a Cancer Dependency Map. Cell. 2017; 170: 564-576, e16. https://doi.org/10.1016/j.cell.2017.06.010 [Europe PMC Full Text] [Europe PMC Abstract]
- 9.A genome-wide atlas of co-essential modules assigns function to uncharacterized genes. Nature Genetics. 2021; 53: 638-649. 2021 53:5 https://doi.org/10.1038/s41588-021-00840-z [Europe PMC Full Text] [Europe PMC Abstract]
- 10.Project DRIVE: A Compendium of Cancer Dependencies and Synthetic Lethal Relationships Uncovered by Large-Scale, Deep RNAi Screening. Cell. 2017; 170: 577-592, e10. [Europe PMC Abstract]
- 11.Gene Essentiality Profiling Reveals Gene Networks and Synthetic Lethal Interactions with Oncogenic Ras. Cell. 2017; 168: 890-903, e15. https://doi.org/10.1016/j.cell.2017.01.013 [Europe PMC Full Text] [Europe PMC Abstract]
- 12.Interrogation of Mammalian Protein Complex Structure, Function, and Membership Using Genome-Scale Fitness Screens. Cell Syst. 2018; 6: 555-568, e7. https://doi.org/10.1016/j.cels.2018.04.011 [Europe PMC Full Text] [Europe PMC Abstract]
- 13.Toward an integrated map of genetic interactions in cancer cells. Mol Syst Biol. 2018; 14. https://doi.org/10.15252/msb.20177656 [Europe PMC Full Text] [Europe PMC Abstract]
- 14.A network of human functional gene interactions from knockout fitness screens in cancer cells. Life Sci Alliance. 2019; 2. https://doi.org/10.26508/lsa.201800278 [Europe PMC Full Text] [Europe PMC Abstract]
- 15.Structure-Function Analysis of the THAP Zinc Finger of THAP1, a Large C2CH DNA-binding Module Linked to Rb/E2F Pathways. Journal of Biological Chemistry. 2008; 283: 4352-4363. [Europe PMC Abstract]
- 16.The dystonia gene THAP1 controls DNA double-strand break repair choice. Mol Cell. 2021; 81: 2611-2624, e10. https://doi.org/10.1016/j.molcel.2021.03.034 [Europe PMC Full Text] [Europe PMC Abstract]
- 17.The THAP-zinc finger protein THAP1 regulates endothelial cell proliferation through modulation of pRB/E2F cell-cycle target genes. Blood. 2007; 109: 584-594. [Europe PMC Abstract]
- 18.The DYT6 Dystonia Protein THAP1 Regulates Myelination Within The Oligodendrocyte Lineage. Dev Cell. 2017; 42: 52. https://doi.org/10.1016/j.devcel.2017.06.009 [Europe PMC Full Text] [Europe PMC Abstract]
- 19.THAP1: Role in Mouse Embryonic Stem Cell Survival and Differentiation. Stem Cell Reports. 2017; 9: 92. https://doi.org/10.1016/j.stemcr.2017.04.032 [Europe PMC Full Text] [Europe PMC Abstract]
- 20.Dystonia type 6 gene product Thap1: identification of a 50 kDa DNA-binding species in neuronal nuclear fractions. Acta Neuropathol Commun. 2014; 2. https://doi.org/10.1186/s40478-014-0139-1 [Europe PMC Full Text] [Europe PMC Abstract]
- 21.Abnormalities of motor function, transcription and cerebellar structure in mouse models of THAP1 dystonia. Hum Mol Genet. 2015; 24: 7159-7170. https://doi.org/10.1093/hmg/ddv384 [Europe PMC Full Text] [Europe PMC Abstract]
- 22.Idiopathic torsion dystonia linked to chromosome 8 in two Mennonite families. Ann Neurol. 1997; 42: 670-673. [Europe PMC Abstract]
- 23.Genotype–Phenotype Relations for Isolated Dystonia Genes: MDSGene Systematic Review. Movement Disorders. 2021; 36: 1086-1103. [Europe PMC Abstract]
- 24.Cytosolic Quality Control of Mislocalized Proteins Requires RNF126 Recruitment to Bag6. Mol Cell. 2014; 55: 227. https://doi.org/10.1016/j.molcel.2014.05.025 [Europe PMC Full Text] [Europe PMC Abstract]
- 25.The tumour suppressor protein VHL targets hypoxia-inducible factors for oxygen-dependent proteolysis. Nature. 1999; 399: 271-275. [Europe PMC Abstract]
- 26.Proteins of Newly Isolated Mutants and the Amino-terminal Proline Are Essential for Ubiquitin-Proteasome-catalyzed Catabolite Degradation of Fructose-1,6-bisphosphatase of Saccharomyces cerevisiae. Journal of Biological Chemistry. 1998; 273: 25000-25005. [Europe PMC Abstract]
- 27.The mdm-2 oncogene product forms a complex with the p53 protein and inhibits p53-mediated transactivation. Cell. 1992; 69: 1237-1245. [Europe PMC Abstract]
- 28.The AMBRA1 E3 ligase adaptor regulates the stability of cyclin D. Nature. 2021; 592: 794-798. https://doi.org/10.1038/s41586-021-03474-7 [Europe PMC Full Text] [Europe PMC Abstract]
- 29.AMBRA1 regulates cyclin D to guard S-phase entry and genomic integrity. Nature. 2021; 592: 799-803. 2021 592:7856 https://doi.org/10.1038/s41586-021-03422-5 [Europe PMC Full Text] [Europe PMC Abstract]
- 30.CRL4AMBRA1 is a master regulator of D-type cyclins. Nature. 2021; 592: 789. https://doi.org/10.1038/s41586-021-03445-y [Europe PMC Full Text] [Europe PMC Abstract]
- 31.Bovine Spleen Multicatalytic Proteinase Complex (Proteasome): Replacement of X, Y, and Z Subunits by LMP7, LMP2, and MECL1 and Changes in Properties and Specificity. Journal of Biological Chemistry. 1997; 272: 11824-11831. [Europe PMC Abstract]
- 32.Tissue Distribution of Constitutive Proteasomes, Immunoproteasomes, and PA28 in Rats. Biochem Biophys Res Commun. 2000; 277: 348-354. [Europe PMC Abstract]
- 33.Improving brightness and photostability of green and red fluorescent proteins for live cell imaging and FRET reporting. Scientific Reports. 2016; 6: 1-12. 2016 6:1 https://doi.org/10.1038/srep20889 [Europe PMC Full Text] [Europe PMC Abstract]
- 34.An Integrated Encyclopedia of DNA Elements in the Human Genome. Nature. 2012; 489: 57. https://doi.org/10.1038/nature11247 [Europe PMC Full Text] [Europe PMC Abstract]
- 35.Identification of multiple binding sites for the THAP domain of the Galileo transposase in the long terminal inverted-repeats. Gene. 2013; 525: 84-89. https://doi.org/10.1016/j.gene.2013.04.050 [Europe PMC Full Text] [Europe PMC Abstract]
- 36.Distinct Elements in the Proteasomal 5 Subunit Propeptide Required for Autocatalytic Processing and Proteasome Assembly *. Journal of Biological Chemistry. 2016; 291: 1991-2003. https://doi.org/10.1074/jbc.M115.677047 [Europe PMC Full Text] [Europe PMC Abstract]
- 37.Proteasome Structure and Assembly. J Mol Biol. 2017; 429: 3500. https://doi.org/10.1016/j.jmb.2017.05.027 [Europe PMC Full Text] [Europe PMC Abstract]
- 38.Proteasome subunits differentially control Myeloma cell viability and proteasome inhibitor sensitivity. Mol Cancer Res. 2020; 18: 1453. https://doi.org/10.1158/1541-7786.MCR-19-1026 [Europe PMC Full Text] [Europe PMC Abstract]
- 39.Global protein stability profiling in mammalian cells. Science (1979). 2008; 322: 918-923. [Europe PMC Abstract]
- 40.Hypoxia-inducible Factor 1α (HIF-1α) Protein Is Rapidly Degraded by the Ubiquitin-Proteasome System under Normoxic Conditions: ITS STABILIZATION BY HYPOXIA DEPENDS ON REDOX-INDUCED CHANGES. Journal of Biological Chemistry. 1997; 272: 22642-22647. [Europe PMC Abstract]
- 41.Dystonia-specific mutations in THAP1 alter transcription of genes associated with neurodevelopment and myelin. The American Journal of Human Genetics. 2021; 108: 2145-2158. https://doi.org/10.1016/j.ajhg.2021.09.017 [Europe PMC Full Text] [Europe PMC Abstract]
- 42.A pathogenic DYT-THAP1 dystonia mutation causes hypomyelination and loss of YY1 binding. Hum Mol Genet. 2022; 31: 1096-1104. https://doi.org/10.1093/hmg/ddab310 [Europe PMC Full Text] [Europe PMC Abstract]
- 43.Roles of METTL3 in cancer: Mechanisms and therapeutic targeting. J Hematol Oncol. 2020; 13: 1-15. https://doi.org/10.1186/s13045-020-00951-w [Europe PMC Full Text] [Europe PMC Abstract]
- 44.Mutations in THAP1 (DYT6) in early-onset dystonia: a genetic screening study. Lancet Neurol. 2009; 8: 441-446. https://doi.org/10.1016/S1474-4422(09)70081-X [Europe PMC Full Text] [Europe PMC Abstract]
- 45.Mutations in the THAP1 gene are responsible for DYT6 primary torsion dystonia. Nature Genetics. 2009; 41: 286-288. 2009 41:3 [Europe PMC Abstract]
- 46.THAP1 mutations (DYT6) are an additional cause of early-onset dystonia. Neurology. 2010; 74: 846. https://doi.org/10.1212/WNL.0b013e3181d5276d [Europe PMC Full Text] [Europe PMC Abstract]
- 47.Prevalence of THAP1 sequence variants in German patients with primary dystonia. Movement Disorders. 2010; 25: 1982-1986. [Europe PMC Abstract]
- 48.Novel THAP1 sequence variants in primary dystonia. Neurology. 2010; 74: 229-238. https://doi.org/10.1212/WNL.0b013e3181ca00ca [Europe PMC Full Text] [Europe PMC Abstract]
- 49.Genotype–phenotype correlations in THAP1 dystonia: Molecular foundations and description of new cases. Parkinsonism Relat Disord. 2012; 18: 414-425. https://doi.org/10.1016/j.parkreldis.2012.02.001 [Europe PMC Full Text] [Europe PMC Abstract]
- 50.Identification and functional analysis of novel THAP1 mutations. European Journal of Human Genetics. 2011; 20: 171-175. 2012 20:2 https://doi.org/10.1038/ejhg.2011.159 [Europe PMC Full Text] [Europe PMC Abstract]
- 51.The THAP-Zinc Finger Protein THAP1 Associates with Coactivator HCF-1 and O-GlcNAc Transferase: A LINK BETWEEN DYT6 AND DYT3 DYSTONIAS*. J Biol Chem. 2010; 285: 13364. https://doi.org/10.1074/jbc.M109.072579 [Europe PMC Full Text] [Europe PMC Abstract]
- 52.Biting the hand that feeds: Rpn4-dependent feedback regulation of proteasome function. Biochim Biophys Acta. 2007; 1773: 1599-1604. [Europe PMC Abstract]
- 53.RPN4 is a ligand, substrate, and transcriptional regulator of the 26S proteasome: a negative feedback circuit. Proc Natl Acad Sci U S A. 2001; 98: 3056-3061. https://doi.org/10.1073/pnas.071022298 [Europe PMC Full Text] [Europe PMC Abstract]
- 54.Transcription factor Nrf1 mediates the proteasome recovery pathway after proteasome inhibition in mammalian cells. Mol Cell. 2010; 38: 17-28. https://doi.org/10.1016/j.molcel.2010.02.029 [Europe PMC Full Text] [Europe PMC Abstract]
- 55.Proteasomal degradation is transcriptionally controlled by TCF11 via an ERAD-dependent feedback loop. Mol Cell. 2010; 40: 147-158. [Europe PMC Abstract]
- 56.FOXO4 is necessary for neural differentiation of human embryonic stem cells. Aging Cell. 2013; 12: 518. https://doi.org/10.1111/acel.12067 [Europe PMC Full Text] [Europe PMC Abstract]
- 57.Regulation of PSMB5 Protein and β Subunits of Mammalian Proteasome by Constitutively Activated Signal Transducer and Activator of Transcription 3 (STAT3): Potential Role in Bortezomib-Mediated Anticancer Therapy*. Journal of Biological Chemistry. 2014; 289: 12612-12622. https://doi.org/10.1074/jbc.M113.542829 [Europe PMC Full Text] [Europe PMC Abstract]
- 58.The CCAAT box-binding transcription factor NF-Y regulates basal expression of human proteasome genes. Biochimica et Biophysica Acta (BBA) - Molecular Cell Research. 2012; 1823: 818-825. [Europe PMC Abstract]
- 59.Destructive cleavage of antigenic peptides either by the immunoproteasome or by the standard proteasome results in differential antigen presentation. J Immunol. 2006; 176: 1053-1061. [Europe PMC Abstract]
- 60.Mice completely lacking immunoproteasomes show major changes in antigen presentation. Nature Immunology. 2011; 13: 129-135. 2011 13:2 https://doi.org/10.1038/ni.2203 [Europe PMC Full Text] [Europe PMC Abstract]
- 61.The immunoproteasome and thymoproteasome: functions, evolution and human disease. Nature Immunology. 2018; 19: 923-931. 2018 19:9 [Europe PMC Abstract]
- 62.DYT6 mutated THAP1 is a cell type dependent regulator of the SP1 family. Brain. 2022; 145: 3968-3984. [Europe PMC Abstract]
- 63.Hallmarks of neurodegenerative diseases. Cell. 2023; 186: 693-714. [Europe PMC Abstract]
- 64.Dysregulated proteostasis network in neuronal diseases. Front Cell Dev Biol. 2023; 11. https://doi.org/10.3389/fcell.2023.1075215 [Europe PMC Full Text] [Europe PMC Abstract]
- 65.Optimized libraries for CRISPR-Cas9 genetic screens with multiple modalities. Nature Communications. 2018; 9: 1-15. 2018 9:1 https://doi.org/10.1038/s41467-018-07901-8 [Europe PMC Full Text] [Europe PMC Abstract]
- 66.The THAP domain of THAP1 is a large C2CH module with zinc-dependent sequence-specific DNA-binding activity. Proc Natl Acad Sci U S A. 2005; 102: 6907-6912. https://doi.org/10.1073/pnas.0406882102 [Europe PMC Full Text] [Europe PMC Abstract]
- 67.A glycine-specific N-degron pathway mediates the quality control of protein N-myristoylation. Science. 2019; 365. https://doi.org/10.1126/science.aaw4912 [Europe PMC Full Text] [Europe PMC Abstract]
- 68.The Eukaryotic Proteome Is Shaped by E3 Ubiquitin Ligases Targeting C-Terminal Degrons. Cell. 2018; 173: 1622-1635, e14. https://doi.org/10.1016/j.cell.2018.04.028 [Europe PMC Full Text] [Europe PMC Abstract]
- 69.g:Profiler: a web server for functional enrichment analysis and conversions of gene lists (2019 update). Nucleic Acids Res. 2019; 47: W191-W198. https://doi.org/10.1093/nar/gkz369 [Europe PMC Full Text] [Europe PMC Abstract]
History
- Posted June 13, 2024.
Citations & impact
This article has not been cited yet.
Impact metrics
Alternative metrics
Discover the attention surrounding your research
https://www.altmetric.com/details/164517184
Funding
Funders who supported this work.
Academy of Medical Sciences (1)
Grant ID: SBF007\1000
0 publications