Abstract
The WT1 (Wilms’ tumour 1) gene encodes a zinc finger transcription factor and RNA-binding protein that direct the development of several organs and tissues. WT1 manifests both tumour suppressor and oncogenic activities, but the reasons behind these opposing functions are still not clear. As a transcriptional regulator, WT1 can either activate or repress numerous target genes resulting in disparate biological effects such as growth, differentiation and apoptosis. The complex nature of WT1 is exemplified by a plethora of isoforms, post-translational modifications and multiple binding partners. How WT1 achieves specificity to regulate a large number of target genes involved in diverse physiological processes is the focus of the present review. We discuss the wealth of the growing molecular information that defines our current understanding of the versatility and utility of WT1 as a master regulator of organ development, a tumour suppressor and an oncogene.
Keywords: co-activator, co-repressor, promoter, transcription, Wilms’ tumour 1 (WT1), zinc finger
INTRODUCTION
The WT1 (Wilms’ tumour 1) protein was among the first tumour suppressor genes to be cloned. Research over the last two decades has revealed much about the functions of WT1 in development and disease, but also many surprises. Indeed, it has become clear that WT1 can act as either a tumour suppressor or an oncogene, it is a transcriptional regulator that can either activate or repress genes, and it is also a post-transcriptional regulator. Through these activities WT1 plays critical roles in development, adult tissue maintenance and also as an epigenetic regulator. These diverse activities are at least partly because of the complex nature of the WT1 gene, its products, post-translational modifications and interaction partners. The purpose of the present review is to summarize the growing molecular information that describes WT1 function. We begin with a brief overview of the structural features of WT1, its isoforms and modifications. We will briefly discuss the role of WT1 as a post-transcriptional regulator and its involvement in RNA metabolism. We then primarily focus on the role of WT1 as a dichotomous regulator of transcription, its target genes and known interaction partners.
WT1 and development
The WT1 protein is indispensable for normal development of the genitourinary system and also several other organs and tissues. WT1-null mice are embryonic lethal with complete agenesis of the kidneys, gonads, heart and diaphragm. WT-null mice die because of heart failure caused by thinning of the epicardium where WT1 appears to be required for proliferation of vascular progenitors [1,2]. WT1-null mice also lack the spleen and adrenal glands [2,3]. Later studies determined an important role for WT1 in the development of neuronal tissues. WT1-null mice do not develop olfactory epithelia or retina ganglia, and show decreases in the proliferation of neuronal progenitor cells [4,5]. More recently, WT1 has been shown to play a role in the development of the peripheral taste system [6], suggesting a significant function for WT1 in the formation of sensory tissues.
Many studies have determined the direct and necessary role of WT1 for normal kidney function. The kidney is formed by interactions between the metanephric mesenchyme and ureteric bud. WT1 is expressed in the metanephric mesenchyme, which condenses to form the epithelium of the nephron [7]. In the absence of WT1, these cells undergo apoptosis. In gain-of-function experiments, microinjection of a WT1-expressing plasmid into isolated embryonic kidneys caused normal nephron development [8]. A previous study used siRNA to ablate WT1 expression at specific times in organ explants of the developing kidney [9]. They demonstrated that WT1 is needed at several stages of kidney development: the condensation of the metanephric mesenchyme on to the ureteric bud, and during the MET (mesenchyme–epithelial transition).
How WT1 controls the development of the kidney is not fully clear. The identification of direct WT1 target genes is crucial to fully understand the role of WT1 in development. A recent study by Kreidberg and co-workers coupled ChIP to a mouse promoter microarray (ChIP-chip) and identified several essential kidney development genes including many previously unknown WT1 target genes involved in nephron progenitor differentiation in vivo [10]. Such studies clearly demonstrate the essential transcriptional role of WT1 in kidney development. It is interesting to note that although, in some instances, WT1 causes neuronal progenitor cells to proliferate, in some other tissues such as the kidney it switches cells from proliferation to differentiation. These different biological functions are tissue-context-dependent and might also be isoform specific. Recent work in uncovering WT1 target genes is providing some new insights into how WT1 functions in different tissues.
WT1 and adult tissue maintenance
In the adult kidney, WT1 expression is restricted to a highly specialized cell type: the podocytes that make up the filtration barrier between blood and urinary spaces [11]. Combining WT1-knockout and inducible yeast artificial chromosome transgenic mouse models, a study demonstrated that reduced expression of WT1 results in the disruption of glomerular health causing either crescentic glomerulonephritis or mesangial sclerosis [12]. Moreover, the two podocyte-specific genes Nphs1 (nephrosis 1) and Podxl (podocalyxin) are down-regulated in WT1-knockout mice [12], which underscores the crucial role of WT1 in podocyte maintenance.
Apart from podocytes, WT1 is also expressed in the Sertoli cells in the testes, granulosa cells in the ovaries, mesothelium and approximately 1 % of cells in the bone marrow of adult mice [11,13–16]. A previous study shed new light regarding the need for WT1 in adult tissues: the specific deletion of WT1 in adult mice caused rapid deterioration of multiple tissues in addition to kidney failure [17]. These included atrophy of the spleen and pancreas and failure of red blood cell formation. Furthermore, within 7–9 days of WT1 deletion, a widespread loss of bone and body fat volume was seen. That study further showed that WT1 loss causes a reduction in the expression of IGF1 (insulin-like growth factor 1), which is a known key regulator of homoeostasis and aging [17]. Collectively these studies show that WT1 plays a role(s) in kidney function in adulthood where it is required for podocyte function and kidney homoeostasis. Transgenic mice continue to provide new insights into WT1 function and the use of tissue-specific Cre lines can be extended to reveal further roles for WT1 in adult tissues. Moreover, a detailed analysis will probably reveal the functions of WT1 in more discrete tissues such as the sensory systems.
WT1 and cancer: tumour suppressor and oncogene
WT1 manifests both tumour suppressor and oncogenic activities, but the molecular mechanisms behind these two activities are still not well understood. The function of WT1 as a classic tumour suppressor is linked to the formation of Wilms’ tumours where WT1 was originally identified [18,19]. Wilms’ tumours are a malignancy of the kidneys that occurs in 1 in 10 000 children [18,20]. The tumours are also associated with hereditary genitourinary disorders such as DDS (Denys–Drash syndrome) and Frasier syndrome [21]. In Wilms’ tumours, a WT1 germline mutation can lead to predisposition to the disease. Also, WT1 is inactivated in many Wilms’ tumours causing increased cell survival and proliferation [22]. Several other genes are mutated in Wilms’ tumour such as WTX [Wilms’ tumour on the X; also known as AMER1 (APC membrane recruitment protein 1)], β-catenin and p53 [18,22]. In addition, LOH (loss of heterozygosity) or LOI (loss of imprinting) on chromosome region 11p15 is observed in up to 70 % of the tumours [23,24]. However, the mechanisms by which one or more genetic modifications cause Wilms’ tumour are unknown. A study by Huff and co-workers shed light on a potential mechanism of action of WT1 in Wilms’ tumours [25]. They produced the first mouse model that mimics a subset of human Wilms’ tumours by engineering mice to sustain somatic deletion of WT1 and constitutive Igf2 expression. That study determined that mice with these combinations developed tumours very early. Furthermore, mechanistically, a lack of WT1 blocked mesenchyme differentiation and thus kidney development leading to cell proliferation and Wilms’ tumours [25]. Thus the Wilms’ tumour mouse model presents a powerful tool in the analysis of WT1 in Wilms’ tumour that provides a better understanding of its tumour suppressor roles in tumorigenesis.
Whereas WT1 behaves as a tumour suppressor gene in Wilms’ tumour, a wealth of data on the expression of WT1 in adult tumours suggest a role as an oncogene. WT1 is expressed in tumours derived from tissues in which it is not normally expressed such as the colon, breast, desmoids and brain [18,26–29]. Furthermore, evidence using antisense oligonucleotides against WT1 demonstrated that ablation of WT1 protein led to cell-cycle arrest at the G1 phase and increased apoptosis of breast cancer cells [30]. Interestingly, WT1 is transcriptionally up-regulated under conditions of low oxygen by HIF (hypoxia-inducible factor) as found in a tumour environment [31]. Furthermore, later studies revealed a role for WT1 in angiogenesis and vascularization, which is an important step for tumour growth. WT1 has been shown to be involved in endothelial cell proliferation and migration [32], and more recently WT1 has been shown to regulate the level of anti-angiogenic VEGF (vascular endothelial growth factor) isoforms [33]. The role of WT1 in leukaemia appears to be complex. WT1 is expressed in most acute leukaemias and it is currently used as a marker of poor prognosis and low survival rates [34]. WT1 is also expressed in immature myeloid leukaemias and CML (chronic myelogenous leukaemia) [35]. Collectively these studies strongly suggest an oncogenic role for WT1 in leukaemogenesis. However, studies also show WT1 mutations in a considerable proportion of both adult and child AML (acute myeloid leukaemia) supporting a tumour suppressor role of WT1. The majority of mutations involve insertions, deletions and mis-sense mutations such as those observed in patients with DDS [22].
How does WT1 act as both a tumour suppressor and oncogene? The oncogenic or tumour suppressive effect of WT1 might result from the cellular conditions at specific time of development and how this responds to modifications in or expression of WT1 [22]. This in turn might be explained by the varying context of cell type and cell status at the moment of WT1 alteration, presence of other downstream genetic modifications and/or microenvironment [22]. This places WT1 in the growing group of proteins such as c-Myc that can exhibit either pro- or anti-tumour activities. The generation of mouse models in which WT1 can be selectively overexpressed in specific tissues will probably shed light on the oncogenic activities of WT1. Regarding tumour treatment, peptide vaccines against WT1 are showing promising results in repressing tumour growth in patients with leukaemia, breast and lung cancer [36]. In addition, the treatment of tumour cell lines with cytotoxic drugs leads to proteolysis of WT1 by the serine protease HtrA2 (HtrA serine peptidase 2) [37]. This process could potentially be exploited to target adult tumours where WT1 acts as an oncogene. The emergence of WT1 as a target for cancer therapy necessitates a greater understanding of the molecular mechanisms of WT1 action, how the different forms of WT1 contribute to these activities and functional consequences of mutations within the WT1 gene. Mouse models to study oncogenic WT1 will help in this regard.
WT1 STRUCTURE
WT1 isoforms
The WT1 gene spans approximately 50 kb at chromosome locus 11p13 and was among the first tumour suppressors to be cloned [38–40]. Many functions have been ascribed to WT1. These include acting as a transcriptional activator and a repressor, a tumour suppressor and oncogene, a function(s) in RNA metabolism, and roles in development, tissue maintenance and disease.
This functional complexity of WT1 can, at least in part, be explained by the generation of multiple protein forms derived from the WT1 gene. At least 24 isoforms of WT1 are generated by a combination of alternative translation start sites, alternative RNA splicing and RNA editing. In mammals, exons 5 and 9 are subjected to alternative splicing, generating four different major splice isoforms [41,42]. These isoforms are also the best studied. Alternative splicing of exon 5 gives rise to a 17-amino-acid insertion (+ 17AA) between the proline/glutamine-rich N-terminus and the C-terminal zinc finger domain of WT1 (Figure 1). Alternative splicing at the end of exon 9 results in either inclusion or exclusion of three amino acids, lysine, threonine and serine (KTS), between the third and fourth zinc finger of WT1 (Figure 1). These two isoforms (+KTS and −KTS) are conserved in all vertebrates, from zebrafish to humans, and imbalanced expression of these variants leads to developmental abnormalities. Fish and non-mammalian vertebrates appear to express only these two variations [43]. This suggests that the different KTS isoforms perform different functions and comprise one of the most important variations. Indeed, WT1 can bind both DNA and RNA and this is influenced by the KTS insertion. Numerous studies have demonstrated that WT1 isoforms lacking the KTS insertion bind to DNA more strongly, and act as transcriptional regulators [44]. The gene product that contains the KTS insertion also acts as a transcriptional regulator, but in addition is associated with post-transcriptional processes. The physiological role of the +KTS and −KTS variants has been studied by mouse models that have a specific isoform deleted. Interestingly, the +KTS variants are essential for the male sex determination programme [45]. The gonads of mice lacking WT1 + KTS variants lack expression of male-specific genes such as Sox9 [SRY (sex-determining region Y) box 9] and Mis [Mullerian-inhibiting substance; also known as Amh (anti-Mullerian hormone)] and develop along the female pathway. It also has been shown that lack of WT1 + KTS isoforms specifically leads to defects in the development of the olfactory system [5].
Figure 1. Functional motifs of the Wilms’ tumour 1 protein WT1.
A linear schematic is shown with numbering indicating the amino acids. The alternative splice sites (17AA and KTS) are shown above along with the post-translational modifications (SUMOylation and phosphorylation). Regions of the protein involved in self-association, nuclear export and nuclear acid binding are shown above. The binding sites of some of WT1 cofactors, such as BASP1, HtrA2, WTX, STAT3, p53, Par-4, CBP and SRY, are indicated below the schematic diagram. Lightning bolts indicate HtrA2 cleavage sites in WT1. A, activation domain; R, repression domain; Zn, zinc finger.
Additional isoforms of WT1 also arise from alternative translation start points. They include the use of an upstream CTG start codon or an internal ATG start codon at the end of exon 1 [18]. Another WT1 isoform, AWT1, arises from the use of an alternative promoter that resides within exon 1 [46]. The authors of that study demonstrated that the AWT1 isoform is imprinted in normal kidney and expression is confined to the paternal allele. However, the properties and function of AWT1 remain to be investigated in vivo.
Determining the role(s) of the different WT1 gene products, gene regulation, and their effects in development and disease is likely to remain a major challenge for some time. Expression of single WT1 isoforms in cultured cells is of questionable physiological relevance, and RNAi-mediated depletion of specific WT1 isoforms is, in large part, not possible. Despite these caveats, such analysis can still yield important insights. Indeed, ectopic expression of individual isoforms of WT1 revealed the function of KTS in directing the interaction of WT1 with RNA processing factors. Although transgenic models have shed light on the role of the KTS insertion in WT1, unfortunately they have not been as fruitful with some of the other WT1 isoforms [43,44]. However, it is clear that our understanding of the roles of WT1 in both development and tissue homoeostasis is not yet complete and other mouse models of WT1 isoforms may still provide insights into their function. Indeed, this may be particularly important in studying the potential oncogenic roles for the different isoforms.
The transcriptional regulatory domains of WT1
The primary sequence of WT1 contains a proline- and glutamine-rich region in the N-terminal domain and four zinc fingers at the C-terminus of the C2-H2 ‘Krüppel’ type [48]. WT1 is a dichotomous transcriptional regulator that can either activate or repress transcription. WT1 has domains that function independently to activate or repress transcription when fused to the GAL4 DNA-binding domain. Specifically, the transcriptional repression domain is contained within WT1 residues 71–180 and the transcriptional activation domain between residues 180 and 250 (Figure 1) [47,48]. The N-terminal region of WT1, specifically two distinct domains residing at amino acids 1–45 and 157–253 mediate self-association [49]. A physiological role for WT1 self association into dimers or multidimers is not yet understood. However, self association of WT1 supports the model of a dominant-negative effect of WT1 mutants on WT1 wild-type protein. Indeed DDS is typified by WT1 proteins that are C-terminally truncated (or contain C-terminal point mutations) that render them defective in binding nucleic acids and yet act in a dominant-negative manner [50].
Refined mutagenesis of the WT1 repression domain identified a subregion (residues 71–101) termed the SD (suppression domain) [50]. The SD inhibits transcriptional activation by WT1 in in vitro transcription assays, but does not inhibit basal activator-independent transcription [20,50]. Moreover, the WT1 SD can inhibit a heterologous transcriptional activation domain in cis both in vivo and in vitro [50]. The WT1 SD functions by recruiting a transcriptional co-repressor, BASP1 (brain acid-soluble protein 1), that inhibits transcriptional activation by WT1 [51]. The SD of WT1 is 90 % conserved across species compared with approximately 50 % identity for the whole of WT1 [50]. To date, the SD has not been identified in any other transcription factors.
Post-translational modification of WT1
It has been shown that WT1 can be SUMOylated at two lysine residues within the N-terminus [52] (Lys73 and Lys177; Figure 1). WT1 had previously been shown to interact with the SUMO-1 (small ubiquitin-related modifier-1) E2-conjugating enzyme Ubc9 (ubiquitin-conjugating enzyme 9) [53]. The sites of WT1 SUMOylation lie within the transcriptional repression domain of WT1, but it is not clear if this modification affects the transcriptional regulatory functions of WT1. WT1 conjugates SUMO-1 and, although WT1 associates with nuclear speckles, the disruption of WT1 SUMOylation does not appear to affect its subnuclear localization [44,52]. Thus it is probable that SUMOylated WT1-binding partners might help to retain WT1 within the speckles. Indeed the WT1-binding partner BASP1 is also SUMOylated, but by SUMO-3 and not SUMO-1 [54]. This raises the potential for the differential regulation of WT1 and BASP1 by SUMOylation. However, it remains to be determined how SUMOylation of WT1 affects its function or association with its cofactors. A more detailed analysis of WT1 SUMOylation in developing tissues or cell line models of differentiation may shed further light on this post-translational modification.
WT1 can also be phosphorylated by PKA (protein kinase A) or PKC (protein kinase C) at Ser365 and Ser393 in the second and third zinc fingers respectively (Figure 1) [55,56]. Phosphorylation of WT1 blocks the ability of WT1 to bind to DNA. This, in turn, affects transcription and decreases transcriptional repression activity in vivo, but leaves the RNA-binding activity intact [55]. The authors also reported that the localization pattern of WT1 in the nucleus is altered by phosphorylation resulting in the cytoplasmic retention of WT1. These findings strongly suggest that phosphorylation negatively regulates the transcriptional role of WT1 by sequestration of WT1 protein in the cytoplasm and inhibition of the WT1 DNA-binding activity [55]. It will be interesting to test if phosphorylation of WT1 affects its post-transcriptional functions or localization to the polysomes (see below), perhaps by modulating interactions between WT1 and cytoplasmic proteins. Thus post-translational modifications represent ways in which the activity of WT1 can be modified. The existence of other WT1 post-translational modifications, such as acetylation or methylation, has yet to be determined. To date, resource studies utilizing global proteomic analysis of post-translational modifications have not used cell types that express WT1. With the number of such proteome-wide analyses growing it is probable that future works will incorporate WT1-expressing cells.
WT1 AS A DICHOTOMOUS REGULATOR OF TRANSCRIPTION
WT1 target genes: growth and development
WT1 functions as a transcriptional regulator that activates or represses a large variety of target genes. The identification of WT1 target genes is an ongoing and difficult process due to the isoform- and context-specific roles of WT1. Many potential target genes of WT1 have been identified; however, it is not fully understood which genes are the biological targets of WT1 and how WT1 orchestrates their regulation in development, tissue homoeostasis or disease. WT1 target genes can be thematically grouped. WT1 can induce expression of genes involved in organ development and the MET. Similarly, WT1 regulates growth factor genes and their receptors. Depending on the cellular context, WT1 can also regulate genes involved in apoptosis. Emerging target genes demonstrate a transcriptional role for WT1 in cytoskeleton organization and cell adhesion, actin biogenesis, cell signalling such as through MAPK (mitogen-activated protein kinase) and WNT, and epigenetic regulation. Table 1 presents a list of all the proposed WT1 target genes, the methods of validation and the organisms in which they were identified.
Table 1.
WT1 target genes
(a) Growth and development | |||
---|---|---|---|
| |||
Target gene | Organism(s) | Method of validation | Reference(s) |
| |||
CXXC5(WID) | Human, mouse | ChIP-chip, ChIP, reporter assay, organ culture morpholino | [10,80] |
BMP7 | Mouse | ChIP-chip, ChIP, organ culture morpholino | [10] |
Pax2 | Mouse, human | ChIP-chip, ChIP, DNase I footprinting, reporter assay | [10,57] |
Sall1 | Mouse | ChIP-chip, ChIP, reporter assay, organ culture morpholino | [10] |
Hey1 | Mouse | ChIP-chip, ChIP, reporter assay, organ culture morpholino | [10] |
Egr3 | Mouse | ChIP-chip, ChIP | [10] |
Fyn | Mouse | ChIP-chip, ChIP | [10] |
Hoxd4 | Mouse | ChIP-chip, ChIP | [10] |
Sox11 | Mouse | ChIP-chip, ChIP, reporter assay, organ culture morpholino | [10] |
Pbx2 | Mouse | ChIP-chip, ChIP, reporter assay, organ culture morpholino | [10] |
Zfr | Mouse | ChIP-chip, ChIP | [10] |
Hhat | Mouse | ChIP-chip, ChIP | [10] |
Arnt2 | Mouse | ChIP-chip, ChIP | [10] |
Erbb2 | Mouse | ChIP-chip, ChIP | [10] |
Smad7 | Mouse | ChIP-chip, ChIP | [10] |
Pbx1 | Mouse | ChIP-chip | [10] |
Pixnb1 | Mouse | ChIP-chip, ChIP, reporter assay, organ culture morpholino | [10] |
BMP4 | Mouse | ChIP-chip, ChIP, qRT-PCR | [6,10] |
Smad4 | Mouse | ChIP-chip, ChIP | [10] |
Smad3 | Mouse | ChIP-chip, ChIP | [10] |
Vegfa | Mouse | ChIP-chip, ChIP | [10] |
PTIP | Mouse | ChIP-chip, ChIP | [10] |
Sulf2 | Mouse | ChIP-chip, ChIP | [10] |
Smad6 | Mouse | ChIP-chip, ChIP | [10] |
Smo | Mouse | ChIP-chip, ChIP | [10] |
ER1 | Human | Reporter assay, ChIP | [135] |
Amhr2 | Mouse | Microarray, EMSA, reporter assay, ChIP | [70] |
IFI16 | Human | Microarray, reporter assay, ChIP | [78] |
VEGF | Human | EMSA, reporter assay, ChIP | [136,137] |
EPO | Human | EMSA, reporter assay, ChIP | [138] |
NTRK2 (TrkB) | Human | EMSA, reporter assay, ChIP | [74] |
IGF1R | Human, Chinese hamster | EMSA, reporter assay, ChIP | [54,65,107,139–141] |
Spry1 (sprouty 1) | Mouse | Reporter assay, ChIP, EMSA | [73] |
Amh (MIS) | Mouse | ChIP, gel shift | [75] |
POU4F2 | Human | Reporter assay | [142] |
Sf1 | Mouse | DNase I footprinting, EMSA, reporter assay | [143] |
CCNE | Human, mouse | Reporter assay, EMSA, ChIP | [131,144] |
Ccng1 | Mouse | Microarray | [145] |
Igfbp4 | Mouse | Expression microarray | [145] |
VDR | Human | Reporter assay, EMSA, ChIP | [127,131,145,146] |
IGF1 | Human | Reporter assay | [140] |
AREG | Human, mouse | DNase I footprint, reporter assay, EMSA, qRT-PCR, ChIP, microarray | [68,76,127,128,131] |
EREG | Human, mouse | Microarray, qRT-PCR, reporter assay, ChIP | [76] |
Amh (MIS) | Human | Reporter assay, EMSA | [117] |
HBEGF | Human, mouse | Microarray, reporter assay, ChIP | [76] |
IL11 | Human, mouse | Microarray, reporter assay | [76] |
CX3CL1 | Human, mouse | Microarray, ChIP, reporter assay | [76] |
IGFII | Mouse | DNase I footprinting, gel mobility-shift assay, reporter assay | [66,88,147–149] |
INSR | Human | Reporter assay, DNase I footprinting, gel mobility-shift assay | [150–152] |
MDK | Human | Gel mobility-shift assay, DNase I footprint, reporter assay | [153] |
Sdc1 | Mouse | Gel mobility-shift assay, DNase I footprint, reporter assay | [58] |
NOV (NovH) | Human | RNase protection assay, primer extension, reporter assay | [67] |
EGFR | Human | EMSA, reporter assay, DNase I footprinting | [64] |
RARA | Human | Gel mobility-shift assay, reporter assay | [63] |
LOC100766733 (Inhibin-α) | Chinese hamster | Reporter assay, RNase protection assay | [154] |
TGFB | Monkey | EMSA, reporter assay | [62] |
ETS1 | Human | EMSA, reporter assay, ChIP | [32,127,131] |
PDGFA | Human, mouse | Gel mobility-shift assay, reporter assay | [53,61,155,156] |
RBBP7 (RbAp46) | Human | Suppression subtractive hybridization PCR | [157] |
SLC6A6 (TauT) | Human | Reporter assay, EMSA | [158] |
WT1 | Human | Reporter assay | [159,160–162] |
CORO1B | Mouse | ChIP, reporter assay | [163] |
Snai2 (Slug) | Mouse | ChIP, reporter assay | [164] |
Inha | Mouse | ChIP, reporter assay | [165] |
(b) Differentiation | |||
| |||
Target gene | Organism(s) | Method of validation | Reference(s) |
| |||
Egr3 | Mouse | ChIP-chip, ChIP | [10] |
Nab2 | Mouse | ChIP-chip, ChIP | [10] |
Sox11 | Mouse | ChIP-chip, ChIP, organ culture morpholino | [10] |
Scx | Mouse | ChIP-chip, ChIP, organ culture morpholino | [10] |
SNAI2 (SLUG) | Human, mouse | Microarray, ChIP, reporter assay | [76] |
Sema6d | Mouse | ChIP-chip, ChIP | [10] |
Rps6ka3 (Rsk2) | Mouse | ChIP-chip, organ culture morpholino | [10] |
Snai2 | Mouse | ChIP, reporter assay | [166] |
CSF1 | Human | Reporter assay | [167] |
Scel | Mouse | ChIP | [168] |
Sulf1 | Mouse | ChIP | [168] |
HOXA10 | Human | EMSA, reporter assay | [169] |
Nphs1 | Mouse, human | Reporter assay, DNase footprinting, EMSA | [81,82] |
TBXA2R | Human | EMSA, reporter assay, ChlP | [170] |
EpoR | Mouse, human | EMSA, ChIP, reporter assay | [171] |
SRY | Human | Reporter assay, gel mobility-shift assay | [71] |
DAX1 | Monkey, human | Footprinting analysis, reporter assay, mobility-shift assay | [172] |
Podxl | Rat, mouse | EMSA, reporter assay, ChIP | [54,69] |
p21 | Human | Expression analysis | [173] |
c-Myb | Human | Footprinting, EMSA, reporter assay | [174] |
Gαi–2 | Pig | Reporter Assay, gel mobility-shift assay | [175] |
(c) Cytoskeleton organization and cell adhesion | |||
| |||
Target gene | Organism(s) | Method of validation | Reference(s) |
| |||
Col4a1 | Mouse | Reporter assay | [176] |
Cik4a2 | Mouse | Reporter assay | [176] |
Actn1 | Mouse | ChIP-chip, ChIP | [10] |
Zyx | Mouse | ChIP-chip, ChIP | [10] |
Lsp1 | Mouse | ChIP-chip, organ culture morpholino | [10] |
Myo1B | Mouse | ChIP-chip | [10] |
CDH1 | Human, mouse | ChIP, reporter assay, EMSA | [166,177–179] |
ITGA4 | Human | ChIP, EMSA, reporter assay | [180] |
NES | Human | Reporter assay | [181] |
Adamts16 | Mouse | EMSA, ChIP, reporter assay | [182] |
(d) WNT signalling | |||
| |||
Target gene | Organism(s) | Method of validation | Reference(s) |
| |||
DKK2 | Human | ChIP-ChIP, microarray | [79] |
DACT1 | Human | ChIP-ChIP, microarray | [79] |
TBL1X | Human | ChIP-ChIP, microarray | [79] |
CCND2 | Human | ChIP-ChIP, microarray | [79] |
LEF1 | Human | ChIP-ChIP, microarray, ChIP, qRT-PCR | [6,10,79] |
JUN | Human | ChIP-ChIP, microarray | [79] |
NLK | Human | ChIP-ChIP, microarray | [79] |
BTRC | Human | ChIP-ChIP, microarray | [79] |
Wnt4 | Mouse, human | Reporter assay, ChIP | [72,129] |
(e) MAPK signalling | |||
| |||
Target gene | Organism(s) | Method of validation | Reference(s) |
| |||
DUSP16 | Human | ChIP-ChIP, microarray | [79] |
DDIT3 | Human | ChIP-ChIP, microarray | [79] |
MAPKAPK2 | Human | ChIP-ChIP, microarray | [79] |
DUSP6 | Human | ChIP-ChIP, microarray, reporter assay, ChIP | [79,183] |
RRAS2 | Human | ChIP-ChIP, microarray | [10,79] |
DUSP5 | Human | ChIP-ChIP, microarray | [79] |
ERK1 | Human | ChIP-chip | [10] |
(f) Apoptosis | |||
| |||
Target gene | Organism(s) | Method of validation | Reference(s) |
| |||
A1/BFL1 | Human | Reporter assay, ChIP | [86] |
BCL2A1 | Human | Reporter assay, EMSA, RNase protection assay | [92] |
BCL2 | Human | Reporter assay, EMSA, RNase protection assay | [85,92] |
c-MYC | Human, mouse | Reporter assay, microarray, EMSA, ChIP | [54,128,184–186] |
n-MYC | Human | Reporter assay, EMSA | [187] |
JunB | Human, mouse | ChIP, microarray | [37,76,131] |
BAK | Human, mouse | ChIP, EMSA, reporter assay, RNase protection assay | [54,92] |
(g) Epigenetic regulation | |||
| |||
Target gene | Organism(s) | Method of validation | Reference(s) |
| |||
DNMT3a | Human | ChIP, reporter assay | [93] |
Rest | Mouse | ChIP-chip, ChIP | [10] |
Jmjd1a | Mouse | ChIP-chip | [10] |
Jmdj3 | Mouse | ChIP-chip | [10] |
Usp16 | Mouse | ChIP-chip | [10] |
Myst2 (HBo1) | Mouse | ChIP-chip | [10] |
SRPK1 | Human | ChIP, reporter assay | [33] |
(h) Others | |||
| |||
Target gene | Organism(s) | Method of validation | Reference(s) |
| |||
STIM1 | Human | ChIP | [188] |
REN | Human | Gel shift, reporter assay, ChIP | [127,189] |
Nos2 | Rat | Reporter assay | [190] |
MMP9 | Human | ChIP | [191] |
TERT | Human | EMSA, reporter assay | [192] |
MDR1 | Human | EMSA, reporter assay | [193] |
TSP1 | Human | Reporter assay | [194] |
ODC | Human, mouse | Reporter assay, DNase 1 protection and methylation interference assays | [195] |
SLC20A1 | Human, mouse | Microarray, ChIP reporter assay | [76] |
IL1RAP | Human, mouse | Microarray, ChIP reporter assay | [76] |
CD95L | Human | ChIP, reporter assay | [196] |
Many of the candidate target genes underscore the growth-regulatory role of WT1, which guides WT1 in development. For many of the target genes identified soon after the discovery of WT1, the transcriptional effects were determined with transient transfection of reporter assays and therefore, in many cases, it is still not clear if they are bona fide WT1 target genes in vivo. Examples include the developmental regulatory gene PAX2 (paired box 2) that is repressed by WT1 during normal kidney development [57] and the syndecan-1 protein, which also reflects the expression of WT1 during kidney development [58]. Syndecan-1 has been shown to have a role in MET in nephrogenesis [58]. Other examples include many growth factors and their receptors such as IGF1R (IGF1 receptor) [59], PDGF-A (platelet-derived growth factor α) [60,61] TGFβ (transforming growth factor β) [62], Rar-α (retinoic acid receptor-α) [63], EGFR [EGF (epidermal growth factor receptor)] [64], insulin receptor [65], IGF2 [66] and the NovH (nephroblastoma overexpressed gene) gene [67]. For some of these genes, further validation using techniques such as ChIP was later performed. The emergence of DNA microarrays provided a new tool to determine biologically relevant target genes of WT1. An early study used microarrays to show that WT1 transcriptionally activates the amphiregulin promoter [68]. Amphiregulin mirrors the pattern of WT1 protein expression in the developing kidney and can induce tubule branching in kidney organ explants. Using the same approach, Podxl was identified as a target gene [69]. Podocalyxin is expressed in the differentiated and mature podocyte cells of the kidney and is a major structural membrane protein of podocytes. DNA microarrays were also used to show that WT1 is an activator of Amhr2 (anti-Mullerian hormone type 2 receptor) [70]. That study generated new insights into the function of WT1 in early gonad development and sex determination. WT1 might mediate this function through the regulation of the sex-determining gene SRY. WT1 activates the SRY gene and initiates a regulatory gene cascade involved in the male sex determination and differentiation pathway [71].
WT1 target genes that explain its function in development and growth have also been validated by ChIP. Examples include WNT4 (wingless-type MMTV integration site family 4), a stimulator of renal development [72], SPRY1 (sprouty homologue 1) a regulator of receptor tyrosine kinase signalling and kidney development [73], TrkB [tropomyosin receptor kinase B; also known as NTRK2 (neurotrophic tyrosine kinase, receptor, type 2)] a growth factor receptor important for coronary vasculature development [74], ETS1 a stimulator of tumour vascularization via regulation of endothelial cell proliferation and migration [32], and MIS a positive regulator of Mullerian duct regression [75].
Since the detailed mechanisms by which WT1 functions in organogenesis in different tissues remains unclear, significant attention has been directed at identifying and further verifying target genes. New emerging approaches such as genome-wide expression profiling analysis in cells expressing inducible WT1 have identified novel target genes that may be important in organogenesis. One such study identified a number of direct WT1 target genes including the EGF family ligands epiregulin and HBEGF (heparin-binding EGF-like growth factor), the chemokine CX3CL1 [chemokine (C-X3-C motif) ligand 1], and the transcriptional factors SLUG [also known as SNAI2 (snail family zinc finger 2)] and JUNB (jun B proto-oncogene) [76]. The genes were validated further by ChIP and luciferase reporter assays. Additionally, the authors used an in vitro embryonic kidney culture system to demonstrate the physiological significance of the novel target genes. Overexpression of the WT1-regulated growth factors epiregulin, amphiregulin, CX3CL1 and the cytokine IL-11 (interleukin-11) significantly increased ureteric bud branching morphogenesis [76]. Although microarray analysis had been performed before [68], the authors improved the sharpness of the data profile by using tetracycline repressible expression profiles at multiple induction times which significantly reduced ‘noise’ and eliminated stochastic changes [76]. Expression profiling analysis in search for WT1 target genes has also been performed by comparing the induction of WT1 with the repression of WT1 [77]. To do so, the authors have created tetracycline-inducible WT1 (−KTS) isoform and WT1 (+ 17AA, + KTS) isoform in a developmentally relevant cell line HEK (human embryonic kidney)-293, which does not express endogenous WT1, and performed a microarray-based expression profile. They also generated a stable transfectant of the mouse mesonephric cell line M15 that endogenously expresses all WT1 isoforms, to express a full-length antisense WT1 construct that abolished the production of all four WT1 isoforms. That study demonstrated that by examining the complementary overlap gene expression changes caused by WT1 induction and repression, the most pronounced change was seen in genes involved in the mevalonate pathway of cholesterol biosynthesis [77]. Since, the effect of WT1 on its target genes is context-dependent, cell line-dependent and isoform-dependent, the study aimed to delineate physiological relevant targets of WT1 by the induction and repression of all WT1 isoforms. ChIP can be used to further validate that genes involved in cholesterol biosynthesis are direct targets of WT1.
Genome-wide analyses have also been performed to find WT1 target genes involved in Wilms’ tumours. A study compared the microarray profiles between WT1 induction in the Saos-2 osteosarcoma cell line and Wilms’ tumours and identified IFI16 (interferon-inducible 16), a transcriptional modulator, as a WT1 target gene [78]. IFI16 is overexpressed in WT1-replete tumours and loss of IFI16 and WT1 inhibits Wilms’ tumour growth. Both genes have different expression profiles in healthy kidneys, but are aberrantly co-expressed in Wilms’ tumours. That study reveals a pathogenic link between WT1 and IFI16 and identified IFI16 as a target gene that supports cell growth.
A previous study also identified WT1 target genes using Wilms’ tumour cell lines. They manipulated WT1 levels in a Wilms’ tumour-derived CCG99-11 cell line by conditional overexpression and shRNA-mediated knockdown [79]. Target genes regulated by WT1 were identified using gene expression profiling in combination with ChIP followed by microarray analysis (ChIP-chip). The microarray was performed in both GOF (gain of function) andLOF (loss of function) systems of WT1. To prioritize WT1 target genes, they compared genes differentially regulated by WT1 in both systems with genes identified in ChIP-chip arrays. Genes directly regulated by WT1 were functionally grouped into MAPK signalling, axon guidance and Wnt pathways, (see Table 1) [79]. As nine target genes were involved in Wnt signalling the authors concluded that one essential role of WT1 in development and/or disease is to regulate the Wnt signalling pathway. In addition, it is significant that the authors used both GOF and LOF of WT1 to identify direct targets. The loss of function of endogenous WT1 leads to loss of all WT1 isoforms and identifies those targets bound by all isoforms, whereas only the WT1 — KTS isoform was induced in the GOF [79]. For the latter, the study could not identify genes that are directly regulated by any of the other WT1 isoforms. Nevertheless, the comparison of genes differentially regulated by WT1 in either GOF or LOF genes with genes identified in ChIP-chip arrays led to identification of new direct targets. Specifically, WT1 regulates the Wnt pathway genes and this might partly explain its function in development.
Many of the efforts to identify WT1 target genes have been dependent on the use of immortalized cell lines and for some identified target genes a biological function in vivo has not been clarified. A previous study sought to identify bona fide WT1 target genes involved in renal development by using a genome-wide ChIP-chip approach using chromatin obtained from embryonic kidneys [9]. Many of the WT1 target genes identified were further validated by direct ChIP assays also using chromatin from embryonic kidneys [10] (see Table 1). Moreover, the target genes were further validated biologically in a novel modified WT1 morpholino LOF model in embryonic kidney explants. Therefore, to gain insight into the role of WT1 during kidney development, the authors used a novel modified antisense ‘vivo-morpholino’ delivery system to examine the effects of WT1 knockdown in kidney explants. Essential kidney development genes previously identified previously such as BMP7 (bone morphogenetic protein 7), PAX2 and SALL1 (spalt-like transcription factor 1) were found in that study. In addition, many novel WT1 target genes, such as LSP1 (lymphocyte-specific protein 1), PBX2 (pre-B-cell leukaemia homeobox 2), PLXDC2 (plexin domain-containing 2), SCX (scleraxis homologue), SOX11 and RPS6KA3 (ribosomal protein S6 kinase, 90 kDa, polypeptide 3), involved in nephron progenitor differentiation in vivo were identified. The authors conclude that this set of target genes clarifies the renal agenesis caused by the absence of WT1. Interestingly, WT1 target genes were also implicated in actin cytoskeleton organization, actin biogenesis, cell–cell signalling, cell adhesion, TGFβ/BMP signalling genes, Wnt signalling and MAPK pathway genes. In summation, by applying ChIP-chip to biological approaches, the study has identified many genes that are co-expressed with WT1 in nephron progenitor cells and explain the critical role of WT1 in nephron progenitor during kidney development in vivo [10].
WT1 target genes: differentiation
WT1 plays a central role in the control of differentiation. In the genome-wide analysis performed by Kreidberg and co-workers, the WT1 target genes identified included several involved in actin cytoskeleton organization and biogenesis, cell adhesion, and cell–cell signalling [10]. Specifically, MYO1B (myosin IB), LSP1 and ACTN1 (actinin α1) were found to be WT1 target genes expressed during kidney development and involved in cytoskeletal interactions. Previously, two transcriptome profiling reports during pre-implantation of mouse development found a requirement for the expression of genes involved in cytoskeletal interactions before major differentiation events [10]. Elevated expression of actin cytoskeleton genes and cell adhesion might be needed to facilitate the morphological changes that enable blastomeres to undergo differentiation [10]. Thus similar genes might be obligatory to cause the dramatic morphological changes associated with nephron progenitor differentiation around ureteric bud tips. In summation, WT1 function in differentiation can be explained, at least in part, by its role in activating genes involved in cytoskeletal interactions and cell–cell adhesion.
The ChIP-chip analysis in mouse embryonic kidney also identified WT1 novel target genes that might mediate WT1 function during nephron progenitor differentiation in vivo [10]. They include RPS6KA3, which has a known function in neurite differentiation along with SOX11, NAB2 (NGFI-A-binding protein 2) involved in Schwann cell differentiation, EGR3 (early growth response factor 3) involved in cellular growth and differentiation, SEMA6D (sema domain, transmembrane domain and cytoplasmic domain 6D) involved in myocardial patterning, SCX involved in heart valve and Sertoli cell differentiation, and CXXC5 (CXXC finger protein 5) which has a dual role in kidney development [80] and myelopoietic progenitor differentiation [10]. These target genes were identified as WT1 target genes in embryonic kidney tissues. Specifically, WT1 target genes, SOX11, SCX, RPS6KA3 and CXXC5, were not only co-expressed with WT1 in nephron progenitors in kidney tissues, but WT1 vivo-morpholino treatment of embryonic kidney explants resulted in reduced expression of these target genes [10]. WT1 morphants exhibited specific reductions in the expression of the target genes above in nephron progenitors or metanephric mesenchyme. Thus this genome-wide study identified novel kidney genes that explain WT1 function in nephron progenitors in vivo.
Other examples of candidate genes that mediate the function of WT1 in differentiation are Podxl and Nphs1 [69,81,82]. WT1 regulates the expression of the major glomerular podocyte membrane protein podocalyxin. Inducible expression of WT1 leads to induction of podocalyxin and supports a role for WT1 in activation of the glomerular differentiation programme [69]. Nphs1 is another WT1 target gene and crucial component of podocytes that is required for glomerular filtration [81,82].
WT1 target genes: apoptosis
Several genes that regulate apoptosis such as Bak (BCL2-antagonist/killer), Bcl2 (B-cell CLL/lymphoma 2), Bcl2A1, c-Myc and JunB are WT1 target genes [83,84]. High levels of Bc1-2 have been reported in sporadic Wilms’ tumours that express elevated wild-type + 17AA isoform [85]. Furthermore, induction of WT1 in murine myeloblast cells leads to up-regulation of another Bcl2 family member A1/BFL1 [86]. The anti-apoptotic gene A1/BFL1 is highly expressed in high-risk leukaemias along with WT1. WT1 also mediates an anti-apoptotic role through the transcriptional control of cell-surface receptors such as EGFR or IGF1R, that mediate survival signals [64,87,88]. Thus, in at least some cellular contexts, WT1 elicits oncogenic potential through inhibition of apoptosis.
The anti-apoptotic function of WT1 is also evident in development. WT1-null mice display an increase in apoptosis of metanephric mesenchyme, which leads to complete agenesis of the kidneys [1]. Furthermore, a study that used organ explants of the developing kidney to monitor the effects of WT1 ablation by siRNA, demonstrated that WT1 loss causes the metanephric mesenchyme to undergo apoptosis [9]. In addition, inhibition of WT1 expression by RNA interference in leukaemic cell lines also leads to apoptosis [89]. Underscoring an anti-apoptotic role for WT1, it has been found that expression of WT1 inhibits p53-mediated apoptosis through binding, stabilization and inactivation of p53 function [90].
Pro-apoptotic effects are observed when WT1 is overexpressed in cell lines such as U2OS, Hep2B and Saos-2 [64,91]. Osteosarcoma cell lines expressing WT1 under an inducible tetracycline-regulated promoter mediate apoptosis through reduced synthesis of the EGFR [64]. Also, WT1 can negatively regulate growth factor receptors such as EGFR and the insulin receptor, repressing survival signals and promoting death [91]. WT1 can also directly induce apoptosis through the regulation of the pro-apoptotic gene BAK [92]. That study demonstrated that expression of wild-type WT1 blocked cellular proliferation and DNA synthesis causing a rapid induction of apoptosis. The physiological conditions under which WT1 promotes apoptosis are not yet clear, but it is very probable that the tumour suppressor functions of WT1 include the induction of apoptosis pathways. The new mouse models of Wilms’ tumour might prove valuable in shedding further light on this aspect of WT1 function in vivo.
WT1 target genes: epigenetic regulation
Recent studies have identified the products of WT1 target genes as being involved in the epigenetic regulation of gene expression. A recent study by Malik and co-workers demonstrated that WT1 transcriptionally regulates DNMT3A (DNA methyltransferase 3A) and WT1 status influences genome-wide promoter methylation [93]. Overexpression of WT1 in Wilms’ tumour cell lines leads to an increase in DNA methylation at several promoters and thus silencing of gene expression. DNMT3A has diverse and complex functions in facilitating transcription of genes by non-promoter CpG island methylation and by demethylation of DNA. In addition, a study by Kreidberg and co-workers using a genome-wide ChIP approach identified several WT1 target genes encoding proteins that modify chromatin [10]. Examples include Jmjd1a [KDM3A (lysine-specific demethylase 3A)], Jmdj3, REST (RE1-silencing transcription factor), Usp16 (ubiquitinspecific peptidase 16; also known Ubp-M) and Myst2 (histone acetyltransferase 2) [94]. Further study of how WT1 regulates these genes, and the interplay with direct interactions between WT1 and chromatin and DNA modification complexes, will provide insights into the epigenetic effects of WT1.
ROLE OF WT1 AS A POST-TRANSCRIPTIONAL REGULATOR
WT1 interacts with splicing factors
Although the present review focuses on the transcriptional role of WT1, there is a wealth of evidence describing the role of WT1 in RNA metabolism. However, the physiological relevance and function of WT1 as a regulator of post-transcriptional processes is still not clear. It been proposed that WT1 −KTS isoforms function optimally as transcriptional factors, whereas the + KTS isoforms might act post-transcriptionally [95]. WT1 + KTS isoforms were found to localize with Sm antigens which are a major snRNP (small nuclear ribonucleoprotein) constituent, in nuclear speckles [96]. The presence of WT1 in speckles is a characteristic shared by splicing factors [96]. WT1 + KTS was found to interact with the splicing factor U2AF65 and was incorporated into spliceosomes. These early findings provided the first evidence of a potential new role for WT1 in pre-mRNA processing [97]. In addition, WT1 + KTS associates with the splicing factor WTAP (WT1-associated protein) and they both localize in the speckles [98]. WT1 + KTS also interacts with the RNA-binding protein and splicing factor RBM4 (RNA-binding motif protein 4). Moreover, WT1 + KTS is able to inhibit the effect of RBM4 on alternative splicing in vivo [99]. Thus, WT1 may modulate alternative splicing through its interactions with splicing factors.
WT1 binding to mRNA
Early studies by Ward and co-workers revealed that WT1 can bind to specific RNA sequences [100]. Both WT1 and its closely related EGR1 can bind to the same DNA sequences of the Igf2 gene. However, WT1, but not EGR1, can bind to Igf2 exonic RNA sequences through its zinc fingers. Specifically, WT1 zinc finger 1, which is not found in EGR1, is important for RNA binding [100]. That zinc finger 1 is critical for RNA binding was also confirmed in vivo in Xenopus oocytes [101]. RNA sequences bound to WT1 were also identified by SELEX (systematic evolution of ligands by exponential enrichment), an in vitro selection procedure that enriches RNA sequences bound by RNA-binding proteins [102]. Interestingly, WT1 associates with actively translating polysomes [103]. Specifically, the WT1 + KTS isoform, but not WT1 − KTS, increases the levels of an unspliced RNA retaining an intron and promotes the association of this RNA with polyribosomes. This study provides a link between the role for WT1 in RNA metabolism and translation. Further studies are needed to fully understand the latter.
Following on from the RNA-binding studies, a search began for candidate mRNA targets [95]. WT1 + KTS isoform was found to bind ACTN1 mRNA via its zinc finger domain [95]. ACTN1 is the first likely physiological mRNA candidate of WT1. Indeed, a previous study also identified actin as a novel interaction partner of WT1 [104]. That study further determined that perturbation of actin influences the cell dynamic properties leading to movement of WT1 off the polysome fractions and cancelled nucleocytoplasmic shuttling of WT1. Interestingly, actin perturbation also changed the WT1 DNA- and RNA-binding abilities [104]. These findings add to the role of WT1 in RNA metabolism in response to actin cytoskeletal changes. Although these studies support a role for WT1 in RNA metabolism, the physiological relevance of a post-transcriptional role of WT1 remains to be confirmed. Of note, a study determined that WT1 + KTS, but not WT1 − KTS, up-regulated Mash1 [also known as Ascl1 (achaete–scute complex homologue 1)] mRNA and protein post-transcriptionally [5]. The authors conclude that WT1 + KTS, which has a role in mRNA processing, acts upstream of Mash1 to promote the normal development of the olfactory system. Many other mRNAs might be regulated by WT1 + KTS in vivo and future work will hopefully shed more light on the post-transcriptional roles of WT1 in development and disease.
THE INTERACTION PARTNERS OF WT1 DNA-BINDING PROTEINS
Several proteins have been identified as WT1 interaction partners, which we have separated into five categories (Table 2). WT1-interacting partners in the first two categories are involved in transcriptional regulation, whereas members of the last three are involved in post-transcriptional regulation, proteolysis of WT1 and epigenetic regulation (Table 2). The interacting partners of WT1 involved in RNA metabolism and processing are discussed in the section above. As transcriptional regulators, members of the first two categories are the DNA-binding transcriptional factors and cofactors that modulate transcription. One of the early studies identified that WT1-mediated transcriptional activation was inhibited by a dominant-negative effect of WT1 mutants identified in patients with DDS that could not bind DNA [105]. In the same study, it was shown that WT1 was able to homodimerize by using the first 180 amino acids of WT1 [82,105]. Thus WT1 is able to self-associate and the association of mutant with wild-type WT1 may be responsible for the dominant-negative activity of mutant WT1 in disease.
Table 2.
WT1-interacting partners
(a) DNA-binding transcription factors | ||
---|---|---|
| ||
Protein | WT1-interaction domain | Reference(s) |
| ||
STAT3 | Residues 1–281 | [113] |
ERα | Not determined | [139] |
SREBP | Zinc fingers | [77] |
Pax2 | Residues 1–242 | [115] |
SRY | Zinc fingers | [116] |
p53 | Zinc fingers (1–2) | [106,107,110] |
BMZF2 | Zinc fingers | [197] |
p73 | Zinc fingers | [108] |
p63 | Not determined | [108] |
Hsp70 | N-terminus | [114] |
TFIIB | Activation domain (residues 180–250) | [50] |
SF1 | N-terminus | [117] |
TBP | Residues 245–297 | [122] |
WT1 | N-terminus | [49] |
ZNF224 | Zinc fingers (−KTS) | [198,199] |
ZNF255 | Zinc fingers | [197,198] |
(b) Transcriptional co-regulators | ||
| ||
Protein | WT1-interaction domain | Reference(s) |
| ||
PINCH-1 | Zinc fingers | [118] |
WTX | Zinc fingers | [119] |
WTIP | Not determined | [200] |
IFI16 | Zinc fingers | [78] |
CITED2 | Zinc fingers | [123] |
BASP1 | Suppression domain (residues 71–101) | [51] |
FHL2 | Activation domain (residues 182–250) | [201] |
CBP | Zinc fingers | [124] |
Par-4 | Zinc fingers, + 17AA | [121,122] |
Ciao1 | Zinc fingers | [202] |
(c) RNA metabolism and processing factors | ||
| ||
Protein | WT1-interaction domain | Reference |
| ||
Actin | Not determined | [104] |
snRNP-U | Zinc fingers | [203] |
PSF | Not determined | [95] |
RBM4 | Zinc fingers (+ KTS) | [99] |
ACTN1 mRNA | Zinc fingers (+ KTS) | [95] |
WTAP | Zinc fingers | [99] |
U2AF65 | Zinc fingers (3–4) | [97] |
Splicing complexes (U2-B, U1-70K and coilin) | Not determined | [96] |
(d) Epigenetic factors | ||
| ||
Protein | WT1-interaction domain | Reference |
| ||
DNMT1 | Not determined | [125] |
PcG complex | Not determined | [125] |
Menin | Not determined | [125] |
(e) Proteolytic factors | ||
| ||
Protein | WT1-interaction domain | Reference |
| ||
HtrA2 | Suppression domain (residues 71–101) | [37] |
Ubc9 | N-Terminus | [53] |
(f) Others | ||
| ||
Protein | WT1-interaction domain | Reference |
| ||
E1B55K | Zinc fingers | [204] |
HCMV-1E2 | Zinc fingers | [205] |
Other DNA-binding transcription factors that interact with WT1 include p53 and p73, which have been shown to compete with WT1 in transcriptional regulation [106–108]. Specifically, WT1 interacts with p53 through its zinc fingers and through this interaction WT1 stabilizes p53 and enhances p53-mediated apoptosis by ultraviolet radiation (Figure 2A) [109]. Moreover, p53 interaction can switch the role of WT1 from an activator of transcription to a repressor (Figure 2B). The other member of the p53 family of products, p73, also binds to WT1 through the zinc fingers and reduces WT1 transcriptional activity and DNA binding by WT1 (Figure 2D) [109]. Work in mice suggests that p53 and WT1 can co-operate in tumorigenesis [110] and p53 andWT1 have been shown to associate at the Igf1r and Podxl promoters [107,111]. ThusWT1 binding to p53-like proteins links WT1 with the stress response, development and tumorigenesis through p53. A recent study using Wilms’ tumour cell lines revealed that mutant WT1 proteins antagonize transcriptional repression mediated by p53 [112]. Whether this effect required recruitment of WT1 to the promoter region by p53 is not clear, but suggests a gain of function by WT1 mutant proteins that lack a functional DNA-binding domain.
Figure 2. DNA-binding transcription factors that associate with WT1.
Mechanisms of action of complexes formed by DNA-binding transcription factors and WT1 are shown. The cofactors lead to either gene activation (left-hand panels) or repression (right-hand panels) by WT1. The small yellow circle in (I) indicates that the interaction is dependent on the splice isoform of WT1.
In relation to tumorigenesis, WT1 interacts with STAT3 (signal transducer and activator of transcription 3) and promotes cell proliferation [113]. Interestingly, WT1 and STAT3 interact in a variety of primaryWilms’ tumour cells, raising the possibility that activated STAT3 and WT1 may play a role in tumorigenesis. On the other hand, WT1 association with the inducible chaperone Hsp70 (heat-shock protein 70) causes inhibition of cellular proliferation [114]. WT1 and Hsp70 physically interact in rat embryonic kidney cells and co-localization of WT1 and Hsp70 is evident within podocytes of the developing kidney (Figure 2E). Thus, by inhibiting proliferation, the WT1–Hsp70 association might be important during kidney differentiation.
WT1 has been shown to associate with Pax2, which is also essential during kidney development [115]. WT1 modifies the Pax2 phenotype in mice and it has been proposed that this might require a direct interaction between WT1 and Pax2 [115]. In mammalian gonads, WT1 and SRY genes form a complex together to regulate transcription. SRY uses WT1 as a transcriptional co-activator and the WT1–SRY interaction is important in testis development (Figure 2C) [116]. WT1 interacts with SF1 (splicing factor 1), which is also essential for mammalian gonadogenesis, and SF1, like SRY, uses WT1 as a transcriptional co-activator (Figure 2G) [117]. The characterization of WT1-interacting partners provides new insights that lead to the identification of important pathways in growth and differentiation. The DNA-binding factors ofWT1 and their role in transcriptional activation or repression is shown in Figure 2.
Transcriptional co-regulators of WT1
Members of the second category of WT1-binding factors are proteins that regulate the transcriptional activity of WT1 (Figure 3). PINCH1 [particularly interesting new Cys-His protein 1; also known as LIMS1 (LIM and senescent cell antigen-like domains 1)], an adaptor protein, is a transcriptional regulator expressed in podocytes that interacts with WT1 and represses Podxl gene expression [118]. Another study has identified that the tumour suppressor WTX shuttles to the nucleus and interacts with WT1 and modulates its transcriptional activity. Specifically, WTX enhances the WT1-mediated transcriptional activation of amphiregulin (Figure 3G) [119]. WT1 can act as a transcriptional cofactor for other proteins such as WTIP (WT-interacting protein). WTIP is expressed at the cell surface in cell–cell junctions, but shuttles to the nucleus and interacts with WT1 when podocytes are subjected to damaging agents [120]. WTIP interaction with WT1 in the nucleus leads to transcriptional repression (Figure 3L). Thus WTIP acts as a transcriptional co-repressor ofWT1 after podocyte injury. It has been suggested that WTIP might monitor podocyte damage and modulate WT1 transcriptional activity to help in maintaining podocyte integrity [120]. Whether this involves an intermediary role for cytoplasmic WT1 is not known, but it has become clear that several WT1-interaction partners can be found in the cytoplasm and also at the cell periphery. Further biochemical studies of cytoplasmic WT1 will be needed to shed light on the functions of WT1 outside of direct gene regulation and potential roles in transducing signals within the cell.
Figure 3. Transcriptional co-regulators of WT1.
Transcriptional regulation by WT1 and its transcriptional cofactors are shown, leading to the formation of either an activator complex (left-hand panels) or a repressor complex (right-hand panels). The small yellow circle in (C) indicates that the interaction is dependent on the splice isoform of WT1. Histone modifications are indicated (Ac; H3K27me3).
WT1 contains separate regions that mediate transcriptional activation (A) and repression (R) (Figure 1). The transcriptional activation domain is suppressed by a ten-amino-acid region (SD) within the N-terminus of the repression domain of WT1. The SD interacts with a transcriptional co-repressor, BASP1, that converts WT1 from an activator into a repressor (Figure 3B) [51]. Like WT1, BASP1 is expressed during kidney development and in the adult podocyte. The functional role of WT1 and BASP1 in development is yet to be determined, but WT1 and BASP1 are co-expressed in several developing organs and tissues [51]. The mechanisms of action of the WT1–BASP1 complex are discussed below.
Par-4 (prostate apoptosis response-4) was initially identified as a WT1-binding protein in a yeast two-hybrid screen [121]. Par-4 interactswith the zinc fingers ofWT1 resulting in the repression of transcription at WT1 target genes (Figure 3D). Par-4 can engage in an interaction with another region of WT1 containing the 17AA-encoding alternatively spliced exon (Figure 3C) [122]. The 17AA region acts as an autonomous transcriptional activation domain that is dependent on a specific interaction with Par-4. Transcriptional activation by the WT1 + 17AA isoform through engagement with Par-4 is one of the mechanisms by which WT1 can inhibit apoptosis. Thus Par-4 can mediate different effects on WT1 function through interaction with distinct domains (and splice isoforms) of WT1.
Like Par-4, Cited2 [Cbp (CREB-binding protein)/p300-interacting transactivator, with Glu/Asp-rich C-terminal domain 2] can also function as either a co-activator or a co-repressor of WT1 transcriptional activity, depending on the promoter or cellular context (Figures 3E and 3F). Cited2 interacts with WT1 and stimulates SF-1 gene transcription, whereas it represses the WT1-mediated transcriptional activation of the amphiregulin gene [123]. Another cofactor of WT1 is the transcriptional co-activator and histone acetyl transferase CBP. WT1 binds to CBP via its first two zinc fingers and this association leads to synergistic activation of WT1 target genes (Figure 3A) [124]. Thus the function ofWT1 can be transcriptionally modulated by both co-repressors and co-activators.
Epigenetic factors and WT1
A previous study found that WT1 can associate with epigenetic factors such as menin and DNMT1 [125]. Menin is a tumour suppressor that has a critical role in the control of H3K4me3 (histone H3 trimethylated at Lys4) and gene transcription. Menin represses Pax2 expression, which is one of the early markers of kidney progenitor cells, via the WT1–PcG (polycomb group) complex. Menin does not bind directly to the Pax2 locus, instead it interacts with WT1 and up-regulates WT1 expression, possibly through post-transcriptional regulation, as WT1 mRNA expression was not affected by menin. WT1 then recruits the PcG complex to the Pax2 gene causing transcriptional repression of Pax2 [122]. Furthermore, WT1 interaction with DNMT1 leads to recruitment of DNMT1 to the Pax2 promoter resulting in hypermethylation of CpG in the Pax2 promoter (Figure 3H). Thus menin and DNMT1 act through WT1 to epigenetically repress genes by recruiting the PcG complex and promoting H3K27 (histone H3 Lys27) methylation [125]. These findings suggest that WT1 controls epigenetic regulation by regulating H3K27 histone modification to repress gene transcription.
Proteolytic factors and WT1
We have reported previously that WT1 is a bona fide substrate of the HtrA2/Omi apoptotic protease [37]. HtrA2 binds to the SD of WT1 and cleaves WT1 at multiple sites (Figure 1). Several studies suggest that WT1 acts as an oncogene in tumours primarily through inhibition of apoptosis [83]. The treatment of tumour cell lines with cytotoxic drugs induces HtrA2 activity, which subsequently degrades WT1. This removes WT1 from the promoters of its target genes and leads to changes in gene expression that enhance apoptosis [37]. Thus the cleavage of WT1 by HtrA2 eliminates key anti-apoptotic functions that are mediated by WT1. These findings provide insights into the function of HtrA2 in the control of the oncogenic activities of WT1. Moreover, HtrA2-mediated cleavage of WT1 could be advantageous to adult cancers where WT1 acts as an oncogene. In contrast, the activities of WT1 as a tumour suppressor would be compromised by the induction of HtrA2. Diverse cytotoxic agents, including many drugs used in cancer treatment, can activate HtrA2. Further studies will be required to determine if chemotherapeutic regimes could be optimized to account for the action of HtrA2 on WT1. For example, drugs that are potent inducers of HtrA2 activity would probably be more effective in the treatment of tumours in which WT1 acts as an oncogene.
Ubc9 was found previously to interact with WT1 [53]. The authors suggested a possible link between the ubiquitin–proteasome proteolytic pathway and WT1. However, it was later identified that the primary function of Ubc9 is concerned with SUMOylation. Indeed, WT1 is directly SUMOylated on Lys73 and Lys177 (Figure 1), the former of which lies within the HtrA2- and BASP1-binding site [52]. This raises the possibility that SUMOylation of WT1 might regulate either or both the proteolysis by HtrA2 and transcriptional repression through BASP1. However, further studies are needed to determine the role of SUMOylation in the regulation of WT1 function.
TRANSCRIPTIONAL REGULATION BY THE WT1–BASP1 COMPLEX
The BASP1 protein and its role as a regulator of WT1 transcription function
The transcriptional activation domain of WT1 is subject to regulation by an SD in the N-terminus of WT1 (Figure 1). We identified BASP1 as a transcriptional co-repressor that can block the function of the transcriptional activation domain ofWT1 [51]. BASP1 is expressed in several cell types, but was first identified in neuronal cells where it binds to the cytoplasmic face of the cell membrane through an N-terminal myristoylation motif [126]. At first, BASP1 seemed an unusual candidate to play a role in transcription; however, BASP1 contains a functional bipartite nuclear localization sequence and BASP1 is found exclusively in the nucleus of some cells [20,51,127]. WT1 associates with BASP1 in cells that naturally express both proteins. Moreover, genome-wide analysis has shown that expression of BASP1 in K562 cells leads to transcriptional repression of >90% of WT1 target genes, suggesting that it is likely to be a significant regulator of WT1 transcription function [127].
Since BASP1 can be found in both the nucleus and cytoplasm, this suggests that BASP1 subcellular localization might be an important point of regulation [51]. Moreover, this property of BASP1 might help, at least in part, to explain the variability of WT1 function at target genes in different cells and tissues. WT1 and BASP1 are present in large complexes in cell lines suggesting that several other components may participate in transcriptional repression mediated by WT1 [54]. Indeed, we have recently reported that the transcriptional co-repressor prohibitin is a component of the BASP1 complex and is also required for WT1-mediated transcriptional repression [128].
Function of the WT1–BASP1 complex in growth and differentiation
WT1 and BASP1 are temporally and spatially co-expressed at several sites in the embryo and adult, including the kidney. Analysis of the WT1–BASP1 complex in a model cell line of podocyte differentiation (MPC5) showed that WT1 and BASP1 co-localized at the Bak, c-Myc and Podxl gene promoters in undifferentiated MPC5 cells [54]. Upon differentiation, the Podxl gene was up-regulated, whereas the Bak and c-Myc promoters were down-regulated. The same study also found that BASP1 could be SUMOylated and the SUMOylation of BASP1 was a dynamic process during the differentiation of podocytes. How SUMOylation of BASP1 affects its function as a WT1 transcriptional co-repressor is not yet clear, but may involve a change in the subnuclear distribution of BASP1. Upon differentiation-dependent SUMOylation of BASP1, BASP1 remained bound to the Bak and c-Myc promoters, but dissociated from the Podxl promoter. It is not clear, however, why BASP1 is selective with specific gene promoters of WT1 involved in the differentiation of the podocytes. Interestingly, WT1 and BASP1 can together divert the differentiation of K562 cells from the blood cell lineage to cells with a neuronal-like morphology that express specific neuronal genes and respond to the neurotransmitter ATP [127]. It is tempting to speculate that the WT1–BASP1 complex might co-operate in the formation of neuronal cell types where BASP1 is also highly expressed. This possibility could be explored further by analysis of WT1 and BASP1 in the development of sensory systems that have been shown to be WT1-dependent.
The mechanism of BASP1-mediated transcriptional repression by WT1
A previous study has associated BASP1 with localized histone the modification at the Wnt4 promoter [129]. Although WT1 activates the Wnt4 gene in the kidney, it represses Wnt4 in the epicardium. WT1 activates Wnt4 in the kidney by recruitment of the co-activator CBP, whereas it represses Wnt4 in the epicardium by recruitment of BASP1. The Wnt4 gene locus is flanked by CTCF (CCCTC-binding factor) sites. In the epicardium, the domain between CTCF sites is in a repressive chromatin state through BASP1 recruitment and WT1 depletion causes a flip to an activation state [130]. Thus WT1 represses Wnt4 in a BASP1-dependent chromatin flip–flop.
How does BASP1 mediate transcription repression by WT1? A surprising finding was that BASP1 is required to be myristoylated at its N-terminus to act as a transcriptional co-repressor [131]. The N-terminal myristoylation of BASP1 facilitates its interaction with PIP2 (phosphatidylinositol 4,5-bisphosphate) [126,132–134]. Indeed, chromatin-bound BASP1 contains PIP2 and this is required for the recruitment of HDAC1 (histone deacetylase 1) to the promoter regions of WT1 target genes to mediate transcriptional repression (Figure 3B) [131]. Moreover, we have recently reported that the transcriptional repressor prohibitin forms an integral component of the WT1–BASP1 complex and is required to recruit ATP-dependent chromatin remodelling activities to the promoters [128]. Prohibitin and BASP1 cooperate through PIP2 to repress transcription by recruitment BRG1 (brahma-related gene 1) and HDAC1 to WT1-dependent promoters [128]. How these chromatin remodelling complexes interface with the WT1–BASP1 complex, and the crucial role that phospholipid plays in mediating these interactions, is not yet clear.
CONCLUDING REMARKS
WT1 functions as a transcriptional regulator that represses or activates the expression of numerous target genes. In the past two decades, ChIP experiments and genome-wide analysis, such as microarray and ChIP-chip experiments, have provided a large wealth of information concerning the genes that WT1 regulates. However, many target genes still have to be validated as biological targets ofWT1 and which cellular pathways they regulate in vivo. Elucidating the biological role of the target genes of WT1 using organ culture morpholino systems, gene targeting and transgenic mice will help to further understand the many functions ascribed to WT1 in development and disease.
In addition, much work over the last 20 years has led to a better understanding of WT1 interaction partners and how they mediate the context-specific activities of WT1. Identification of WT1-interacting proteins is an ongoing task and proteomic screens are likely to yield new insights in the coming years. Further studies of epigenetic regulation by WT1 and how this is modulated by its interaction partners will lead to a better understanding of how WT1 establishes gene transcription profiles that regulate developmental and disease processes. Although not the focus of the present review, it is probable that in the next few years we will also learn more about the function of WT1 as an RNA-binding protein. Going forward, once the downstream mediators of WT1 and its cofactors have been characterized by biochemical and cellular approaches followed by the physiological relevance in vivo, the sometimes-confusing functions of WT1 will be resolved and its functions in development and disease will be better understood.
ACKNOWLEDGEMENTS
We thank members of our laboratories for comments on the review and apologize to the colleagues whose work could not be cited owing to space constraints.
Abbreviations:
- ACTN1
actinin α1
- Amhr2
anti-Mullerian hormone receptor 2
- Bak
BCL2-antagonist/killer
- BASP1
brain acid-soluble protein 1
- BCL2
B-cell CLL/lymphoma 2
- BMP
bone morphogenetic protein
- CBP
CREB-binding protein
- Cited2
Cbp/p300-interacting transactivator, with Glu/Asp-rich carboxy-terminal domain 2
- CTCF
CCCTC-binding factor
- CX3CL1
chemokine (C-X3-C motif) ligand 1
- CXXC5
CXXC finger protein 5
- DDS
Denys–Drash syndrome
- DNMT
DNA methyltransferase
- EGF
epidermal growth factor
- EGFR
EGF receptor
- EGR
early growth response factor
- GOF
gain of function
- HDAC1
histone deacetylase 1
- H3K27
histone H3 lysine 27
- Hsp70
heat-shock protein 70
- HtrA2
HtrA serine peptidase 2
- IFI16
interferon-inducible 16
- IGF
insulin-like growth factor
- IGF1R
IGF1 receptor
- JUNB
jun B proto-oncogene
- LOF
loss of function
- LSP1
lymphocyte-specific protein 1
- MAPK
mitogen-activated protein kinase
- MET
mesenchyme–epithelial transition
- MIS
Mullerian-inhibiting substance
- Par-4
prostate apoptosis response-4
- PAX2
paired box 2
- PcG
polycomb group
- PIP2
phosphatidylinositol 4,5-bisphosphate
- RBM4
RNA-binding motif protein 4
- RPS6KA3
ribosomal protein S6 kinase, 90 kDa, polypeptide 3
- SCX
scleraxis homologue
- SD
suppression domain
- SF1
splicing factor 1
- SOX
SRY box
- SRY
sex-determining region Y
- STAT3
signal transducer and activator of transcription 3
- SUMO
small ubiquitin-related modifier
- TGFβ
transforming growth factor β
- Ubc9
ubiquitin-conjugating enzyme 9
- WNT
wingless-type MMTV integration site family
- WT1
Wilms’ tumour 1
- WTIP
WT-interacting protein
- WTX
Wilms’ tumour on the X
REFERENCES
- 1.Kreidberg JA, Sariola H, Loring JM, Maeda M, Pelletier J, Housman D and Jaenisch R (1993) WT-1 is required for early kidney development. Cell 74, 679–691 [DOI] [PubMed] [Google Scholar]
- 2.Moore AW, McInnes L, Kreidberg J, Hastie ND and Schedl A (1999) YAC complementation shows a requirement for Wt1 in the development of epicardium, adrenal gland and throughout nephrogenesis. Development 126, 1845–1857 [DOI] [PubMed] [Google Scholar]
- 3.Herzer U, Crocoll A, Barton D, Howells N and Englert C (1999) The Wilms tumor suppressor gene Wt1 is required for development of the spleen. Curr. Biol 9, 837–840 [DOI] [PubMed] [Google Scholar]
- 4.Wagner KD, Wagner N, Vidal VPI, Schley G, Wilhelm D, Schedl A, Englert C and Scholz H (2002) The Wilms’ tumor gene WT1 is required for normal development of the retina. EMBO J. 21, 1398–1405 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Wagner N, Wagner KD, Hammes A, Kirschner KM, Vidal VP, Schedl A and Scholz H (2005) A splice variant of the Wilms’ tumour suppressor Wt1 is required for normal development of the olfactory system. Development 132, 1327–1336 [DOI] [PubMed] [Google Scholar]
- 6.Gao Y, Toska E, Denmon D, Roberts SGE and Medler KF (2014) WT1 regulates the development of the posterior taste field. Development, doi: 10.1242/dev.105676 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Scholz H and Kirschner KM (2005) A role for the Wilms’tumor protein WT1 in organ development. Physiology 20, 54–59 [DOI] [PubMed] [Google Scholar]
- 8.Gao X, Chen X, Taglienti M, Rumballe B, Little MH and Kreidberg JA (2005) Angioblast-mesenchyme induction of early kidney development is mediated by Wt1 and Vegfa. Development 132, 5437–5449 [DOI] [PubMed] [Google Scholar]
- 9.Davies JA, Ladomery M, Hohenstein P, Michael L, Shafe A, Spraggon L and Hastie N (2004) Development of an siRNA-based method for repressing specific genes in renal organ culture and its use to show that the Wt1 tumour suppressor is required for nephron differentiation. Hum. Mol. Genet 13, 235–246 [DOI] [PubMed] [Google Scholar]
- 10.Hartwig S, Ho J, Pandey P, Macisaac K, Taglienti M, Xiang M, Alterovitz G, Ramoni M, Fraenkel E and Kreidberg JA. (2010) Genomic characterization of Wilms’ tumor suppressor 1 targets in nephron progenitor cells during kidney development. Development 137, 1189–1203 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Armstrong JF, Pritchard-Jones K, Bickmore WA, Hastie ND and Bard JB (1993) The expression of the Wilms’ tumor gene, WT1, in the developing mammalian embryo. Mech. Dev 40, 85–97 [DOI] [PubMed] [Google Scholar]
- 12.Guo JK, Menke AL, Gubler MC, Clarke AR, Harrison D, Hammes A, Hastie ND and Schedl A (2002) WT1 is a key regulator of podocyte function: reduced expression levels cause crescentic glomerulonephritis and mesangial sclerosis. Hum. Mol. Genet 11, 651–659 [DOI] [PubMed] [Google Scholar]
- 13.Walker C, Rutten F, Yuan X, Pass H, Mew DM and Everitt J (1994) Wilms’ tumor suppressor gene expression in rat and human mesothelioma. Cancer Res. 54, 3101–3106 [PubMed] [Google Scholar]
- 14.Rao MK, Pham J, Imam JS, MacLean JA, Murali D, Furuta Y, Sinha-Hikim AP and Wilkinson MF (2006) Tissue-specific RNAi reveals that WT1 expression in nurse cells controls germ cell survival and spermatogenesis. Genes Dev. 20, 147–152 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Pelletier J, Schalling M, Buckler AJ, Rogers A, Haber DA and Housman D (1991) Expression of the Wilms’ tumor gene WT1 in the murine urogenital system. Genes Dev. 5, 1345–1356 [DOI] [PubMed] [Google Scholar]
- 16.Hosen N, Shirakata T, Nishida S, Yanagihara M, Tsuboi A, Kawakami M, Oji Y, Okabe M, Tan B, Sugiyama H et al. (2007) The Wilms’ tumor gene WT1–GFP knock-in mouse reveals the dynamic regulation of WT1 expression in normal and leukemic hematopoiesis. Leukemia 21, 1783–1791 [DOI] [PubMed] [Google Scholar]
- 17.Chau YY, Brownstein D, Mjoseng H, Lee WC, Buza-Vidas N, Nerlov C, Jacobsen SE, Perry P, Berry R, Thornburn A. et al. (2011) Acute multiple organ failure in adult mice deleted for the developmental regulator Wt1. PloS Genet. 7, e1002404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Hohenstein P and Hastie ND (2006) The many facets of the Wilms’ tumour gene, WT1. Hum. Mol. Genet 15, 196–201 [DOI] [PubMed] [Google Scholar]
- 19.Miller-Hodges E and Hohenstein P (2011) WT1 in disease: shifting the epithelial mesenchymal balance. J. Pathol 226, 229–240 [DOI] [PubMed] [Google Scholar]
- 20.Wagner KJ and Roberts SGE (2004) Transcriptional regulation by the Wilms’ tumour suppressor protein WT1. Biochem. Soc. Trans 32, 932–935 [DOI] [PubMed] [Google Scholar]
- 21.Davies R, Moore A, Schedl A, Bratt E, Miyahawa K, Ladomery M, Miles C, Menke A, van Heyningen V and Hastie N (1999) Multiple roles for the Wilms’ tumor suppressor, WT1. Cancer Res. 59, 1747–1750 [PubMed] [Google Scholar]
- 22.Huff V (2011) Wilms’ tumours: about tumour suppressor genes, an oncogene and a chameleon gene. Nat. Rev. Cancer 11, 111–121 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Ogawa O, Eccles MR, Yun K, Mueller RF, Holdaway MDD and Reeve AE (1993) A novel insertional mutation at the third zinc finger coding region of the WT1 gene in Denys–Drash syndrome. Hum. Mol. Genet 1, 203–204 [DOI] [PubMed] [Google Scholar]
- 24.Steenman MJ, Rainier Shirley, Dobry CJ, Grundy P, Horon IL and Feinberg AP (1994) Loss of imprinting of IGF2 is linked to reduced expression and abnormal methylation of H19 in Wilms’ tumour. Nat. Genet 7, 433–439 [DOI] [PubMed] [Google Scholar]
- 25.Hu Q, Gao F, Tian W, Ruteshouser CE, Wang Yaqing, Lazar A, Stewart J, Strong LC, Behringer RR and Huff V. (2011) Wt1 ablation and Ifg2 upregulation in mice result in Wilms’ tumor with elevated ERK1/2 phosphorylation. J. Clin. Invest 121, 174–183 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Koesters R, Linnebacher M, Coy JF, Germann A, Schwitalle Y, Findeisen P and von Knebel Doeberitz M (2004) WT1 is a tumor-associated antigen in colon cancer that can be recognized by in vitro stimulated cytotoxic T cells. Int. J. Cancer 109, 385–392 [DOI] [PubMed] [Google Scholar]
- 27.Loeb DM, Evron E, Patel CB, Sharma PM, Niranjan B, Buluwela L, Weitzman SA, Korz D, and Sukumar S. (2001) Wilms’ tumor suppressor gene (WT1) is expressed in primary breast tumors despite tumor-specific promoter methylation. Cancer Res. 61, 921–925 [PubMed] [Google Scholar]
- 28.Amini Nik S, Hohenstein P, Jadidizadeh A, Van Dam K, Bastidas A, Berry RL, Patek CE, Van der Schueren B, Cassiman JJ and Tejpar S (2005) Upregulation of Wilms’ tumor gene 1 (WT1) in desmoid tumors. Int. J. Cancer 114, 202–208 [DOI] [PubMed] [Google Scholar]
- 29.Oji Y, Suzuki T, Nakano Y, Maruno M, Nakatsuka S, Jomgeow T, Abeno S, Tatsumi N, Yokota A, Aoyagi S et al. (2004) Overexpression of the Wilms’ tumor gene WT1 in primary astrocytic tumors. Cancer Sci. 95, 822–827 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Tuna M, Chavez-Reyes A and Tari AM (2005) HER2/neu increases the expression of Wilms’ tumor 1 (WT1) protein to stimulate S-phase proliferation and inhibit apoptosis in breast cancer cells. Oncogene 24, 1648–1652 [DOI] [PubMed] [Google Scholar]
- 31.Wagner KD, Wagner N, Wellmann S, Schley G, Bondke A, Theres H and Scholz H (2003) Oxygen-regulated expression of the Wilms’ tumor suppressor Wt1 involves hypoxia-inducible factor-1 (HIF-1). FASEB J. 17, 364–366 [DOI] [PubMed] [Google Scholar]
- 32.Wagner N, Michiels JF, Schedl A and Wagner KD (2008) The Wilms’ tumour suppressor WT1 is involved in endothelial cell proliferation and migration: expression in tumour vessels in vivo. Oncogene 27, 3662–3672 [DOI] [PubMed] [Google Scholar]
- 33.Amin EM, Oltean S, Hua J, Gammons MV, Hamdollah-Zadeh M, Welsh GI, Cheung MK, Ni L, Kase S, Rennel ES et al. (2011) WT1 mutants reveal SRPK1 to be a downstream angiogenesis target by altering VEGF splicing. Cancer Cell 20, 768–780 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Inoue K, Sugiyama H, Ogawa H, Nakagawa M, Yamagami T, Miwa H, Kita K, Hiraoka A, Masaoka T, Nasu K et al. (1994) WT1 as a new prognostic factor and a new marker for the detection of minimal residual disease in acute leukemia. Blood 84, 3071–3079 [PubMed] [Google Scholar]
- 35.Miwa H, Beran M and Saunders GF. (1992) Expression of the Wilms’ tumor gene (WT1) in human leukemias. Leukemia 6, 405–409 [PubMed] [Google Scholar]
- 36.Oka Y, Tsuboi A, Taguchi T, Osaki T, Kyo T, Nakajima H, Elisseeva OA, Oji Y, Kawakami M, Ikegame K et al. (2004) Induction of WT1 (Wilms’ tumor gene)-specific cytotoxic T lymphocytes by WT1 peptide vaccine and the resultant cancer regression. Proc. Natl. Acad. Sci. U.S.A 101, 13885–13890 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Hartkamp J, Carpenter B and Roberts SGE (2010) The Wilms’ tumour suppressor protein WT1 is processed by the serine protease HtrA2/Omi. Mol. Cell 37, 159–171 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Haber DA, Buckler AJ, Glaser T, Call KM, Pelletier J, Sohn RL, Douglass EC and Housman DE (1990) An internal deletion within an 11p13 zinc finger gene contributes to the development of Wilms’ tumor. Cell 61, 1257–1269 [DOI] [PubMed] [Google Scholar]
- 39.Call KM, Glaser T, Ito CY, Buckler AJ, Pelletier J, Haber DA, Rose EA, Kral A, Yeger H and Lewis WH (1990) Isolation and characterization of a zinc finger polypeptide gene at the human chromosome 11 Wilms’ tumor locus. Cell 60, 509–520 [DOI] [PubMed] [Google Scholar]
- 40.Gessler M, Poustka A, Cavenee W, Neve RL, Orkin SH and Bruns GA (1990) Homozygous deletion in Wilms tumours of a zinc-finger gene identified by chromosome jumping. Nature 343, 774–778 [DOI] [PubMed] [Google Scholar]
- 41.Gessler M, Konig A and Bruns GA (1992) The genomic organization and expression of the WT1 gene. Genomics 12, 807–813 [DOI] [PubMed] [Google Scholar]
- 42.Haber DA, Sohn RL, Buckler AJ, Pelletier J, Call KM and Housman DE (1991) Alternative splicing and genomic structure of the Wilms tumor gene WT1. Proc. Natl. Acad. Sci. U.S.A 88, 9618–9622 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Hastie ND (2001) Life, sex, and WT1 isoforms – three amino acids can make all the difference. Cell 106, 391–394 [DOI] [PubMed] [Google Scholar]
- 44.Roberts SGE (2005) Transcriptional regulation by WT1 in development. Curr. Opin. Genet. Dev 15, 542–547 [DOI] [PubMed] [Google Scholar]
- 45.Hammes A, Guo JK, Lutsch G, Leheste JR, Landrock D, Ziegler U, Gubler MC and Schedl A (2001) Two splice variants of the Wilms’ tumor 1 gene have distinct functions during sex determination and nephron formation. Cell 106, 319–329 [DOI] [PubMed] [Google Scholar]
- 46.Dallosso AR, Hancock AL, Brown KW, Williams AC, Jackson S and Malik K (2004) Genomic imprinting at the WT1 gene involves a novel coding transcript (AWT1) that shows deregulation in Wilms’ tumours. Hum. Mol. Genet 13, 405–415 [DOI] [PubMed] [Google Scholar]
- 47.Wang ZY, Qiu QQ, Enger KT and Deuel TF (1993) A second transcriptionally active DNA-binding site for the Wilms tumor gene product, WT1. Proc. Natl. Acad. Sci. U.S.A 90, 8896–8900 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Madden SL, Cook DM and Rauscher FJ III. (1993) A structure-function analysis of transcriptional repression mediated by the WT1, Wilms’ tumor suppressor protein. Oncogene 8, 1713–1720 [PubMed] [Google Scholar]
- 49.Holmes GP, Boterashvili S, English M, Wainwright BJ, Licht JD and Little M (1997) Two N-terminal self-association domains are required for the dominant negative transcriptional activity of WT1 Denys–Drash mutant proteins. Biochem. Biophys. Res. Commun 233, 723–728 [DOI] [PubMed] [Google Scholar]
- 50.McKay LM, Carpenter B and Roberts SGE (1999) Regulation of the Wilms’ tumour suppressor protein transcriptional activation domain. Oncogene 18, 6546–6554 [DOI] [PubMed] [Google Scholar]
- 51.Carpenter B, Hill KJ, Charalambous M, Wagner KJ, Lahiri D, James DI, Anderson JS, Schumacher V, Royer-Pokora B, Mann M et al. (2004) BASP1 is a transcriptional cosuppressor for the Wilms’ tumour suppressor protein WT1. Mol. Cell. Biol 24, 537–549 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Smolen GA, Vassileva MT, Wells J, Matunis MJ and Haber DA (2004) SUMO-1 modification of the Wilms’ tumor suppressor WT1. Cancer Res. 64, 7846–7851 [DOI] [PubMed] [Google Scholar]
- 53.Wang ZY, Qiu QQ, Seufert W, Taguchi T, Testa JR, Whitmore SA, Callen DF, Welsh D, Shenk T and Deuel TF (1996) Molecular cloning of the cDNA and chromosome localization of the gene for human ubiquitin-conjugating enzyme 9. J. Biol. Chem 271, 24811–24816 [DOI] [PubMed] [Google Scholar]
- 54.Green LM, Wagner KJ, Campbell HA, Addison K and Roberts SGE (2009) Dynamic interaction between WT1 and BASP1 in transcriptional regulation during differentiation. Nucleic Acids Res. 37, 431–440 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Ye Y, Raychaudhuri B, Gurney A, Campbell CE and Williams BR (1996) Regulation of WT1 by phosphorylation: inhibition of DNA binding, alteration of transcriptional activity and cellular translocation. EMBO J. 15, 5606–5615 [PMC free article] [PubMed] [Google Scholar]
- 56.Sakamoto Y, Yoshida M, Semba K and Hunter T (1997) Inhibition of the DNA-binding and transcriptional repression activity of the Wilms’ tumor gene product, WT1, by cAMP-dependent protein kinase-mediated phosphorylation of Ser-365 and Ser-393 in the zinc finger domain. Oncogene 15, 2001–2012 [DOI] [PubMed] [Google Scholar]
- 57.Ryan G, Steele-Perkins V, Morris JF, Rauscher FJ III, and Dressler GR (1995) Repression of Pax-2 by WT1 during normal kidney development. Development 121, 867–875 [DOI] [PubMed] [Google Scholar]
- 58.Cook DM, Hinkes MT, Bernfield M and Rauscher FJ (1996) Transcriptional activation of the syndecan-1 promoter by the Wilms’ tumor protein WT1. Oncogene 13, 1789–1799 [PubMed] [Google Scholar]
- 59.Werner H, Re GG, Drummond IA, Sukhatme VP, Rauscher FJ III, Sens DA, Garvin AJ, LeRoith D, and Roberts CT Jr (1993) Increased expression of the insulin-like growth factor-I receptor gene, IGFIR, in Wilms’ tumor is correlated with modulation of IGFIR promoter activity by the WT1 Wilms’ tumor gene product. Proc. Natl. Acad. Sci. U.S.A 90, 5828–5832 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Gashler AL, Bonthron DT, Madden SL, Rauscher FJ, Collins T and Sukhatme VP (1992) Human platelet-derived growth factor A chain is transcriptionally repressed by the Wilms’ tumor suppressor WT1. Proc. Natl. Acad. Sci. U.S.A 89, 10984–10988 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Wang ZY, Madden SL, Deuel TF and Rauscher FJ (1992) The Wilms’ tumor gene product, WT1, represses transcription of the platelet-derived growth factor A-chain gene. J. Biol. Chem 267, 21999–22002 [PubMed] [Google Scholar]
- 62.Dey BR, Sukhatme VP, Roberts AB, Sporn MB, Rauscher FJ and Kim SJ (1994) Repression of the transforming growth factor-β 1 gene by the Wilms’ tumor suppressor WT1 gene product Mol. Endocrinol 8, 595–602 [DOI] [PubMed] [Google Scholar]
- 63.Goodyer P, Dehbi M, Torban E, Bruening W and Pelletier J (1995) Repression of the retinoic acid receptor-α gene by the Wilms’ tumor suppressor gene product, WT1. Oncogene 10, 1125–1129 [PubMed] [Google Scholar]
- 64.Englert C, Hou X, Maheswaran S, Bennett P, Ngwu C, Re GG, Garvin AJ, Rosner MR and Haber DA (1995) WT1 suppresses synthesis of the epidermal growth factor receptor and induces apoptosis. EMBO J. 14, 4662–4675 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Werner H, Shen-Orr Z, Rauscher FJ III, Morris JF, Roberts CT Jr. and LeRoith D. (1995) Inhibition of cellular proliferation by the Wilms’ tumor suppressor WT1 is associated with suppression of insulin-like growth factor I receptor gene expression. Mol. Cell. Biol 15, 3516–3522 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Drummond IA, Madden SL, Rohwer-Nutter P, Bell GI, Sukhatme VP and Rauscher FJ III. (1992) Repression of the insulin-like growth factor II gene by the Wilms tumor suppressor WT1. Science 257, 674–678 [DOI] [PubMed] [Google Scholar]
- 67.Martinerie C, Chevalier G, Rauscher FJ. and Perbal B. (1996) Regulation of nov by WT1: a potential role for nov in nephrogenesis. Oncogene 12, 1479–1492 [PubMed] [Google Scholar]
- 68.Lee SB, Huang K, Palmer R, Truong VB, Herzlinger D, Kolquist KA, Wong J, Paulding C, Yoon SK, Gerald W. et al. (1999) The Wilms tumor suppressor WT1 encodes a transcriptional activator of amphiregulin. Cell 98, 663–673 [DOI] [PubMed] [Google Scholar]
- 69.Palmer RE, Kotsianti A, Cadman B, Boyd T, Gerald W and Haber DA (2001) WT1 regulates the expression of the major glomerular podocyte membrane protein podocalyxin. Curr. Biol 11, 1805–1809 [DOI] [PubMed] [Google Scholar]
- 70.Klattig J, Sierig R, Kruspe D, Besenbeck B and Englert C (2007) Wilms’ tumor protein Wt1 is an activator of the anti-MüLlerian hormone receptor gene Amhr2. Mol. Cell. Biol 27, 4355–4364 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Hossain A and Saunders GF (2001) The human sex-determining gene SRY is a direct target of WT1. J. Biol. Chem 276, 16817–16823 [DOI] [PubMed] [Google Scholar]
- 72.Sim EU, Smith A, Szilagi E, Rae F, Ioannou P, Lindsay MH and Little MH (2002) Wnt-4 regulation by the Wilms’ tumour suppressor gene, WT1. Oncogene 21, 2948–2960 [DOI] [PubMed] [Google Scholar]
- 73.Gross I, Morrison DJ, Hyink DP, Georgas K, English MA, Mericskay M, Hosono S, Sassoon D, Wilson PD, Little M. and Licht JD. (2003) The receptor tyrosine kinase regulator Sprouty1 is a target of the tumor suppressor WT1 and important for kidney development. J. Biol. Chem 278, 41420–41430 [DOI] [PubMed] [Google Scholar]
- 74.Wagner N, Wagner KD, Theres H, Englert C, Schedl A and Scholz H (2005) Coronary vessel development requires activation of the TrkB neurotrophin receptor by the Wilms’ tumor transcription factor Wt1. Genes Dev. 19, 2631–2642 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Hossain A and Saunders GF (2003) Role of Wilms tumor 1 (WT1) in the transcriptional regulation of the Müllerian-inhibiting substance promoter. Biol. Reprod 69, 1808–1814 [DOI] [PubMed] [Google Scholar]
- 76.Kim HS, Kim MS, Hancock AL, Harper JC, Park JY, Poy G, Perantoni AO, Cam M, Malik K and Lee SB (2007) Identification of novel Wilms’ tumor suppressor gene target genes implicated in kidney development. J. Biol. Chem 282, 16278–16287 [DOI] [PubMed] [Google Scholar]
- 77.Rae FK, Martinez G, Gillinder KR, Smith A, Shooter G, Forrest AR, Grimmond SM and Little MH. (2004) Analysis of complementary expression profiles following WT1 induction versus repression reveals the cholesterol/fatty acid synthetic pathways as a possible major target of WT1. Oncogene 23, 3067–3079 [DOI] [PubMed] [Google Scholar]
- 78.Kim MK, Mason JM, Li CM, Berkofsky-Fessler W, Jiang L, Choubey D, Grundy PE, Tycko B, and Licht JD (2008) A pathologic link between Wilms tumor suppressor gene, WT1, and IFI16. Neoplasia 10, 69–78 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Kim MK, McGarry TJ, O’Broin P, Flatow JM, Golden AA and Licht JD (2009) An integrated genome screen identifies the Wnt signaling pathway as a major target of WT1. Proc. Natl. Acad. Sci. U.S.A 106, 11154–11159 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Kim MS, Yoon SK, Bollig F, Kitagaki J, Hur W, Whye NJ, Wu YP, Riviera MN, Park JY, Kim HS et al. (2010) A novel Wilms’ tumor 1 (WT1) target gene negatively regulates the WNT signaling pathway. J. Biol. Chem 285, 14585–14593 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Guo G, Morrison DJ, Licht JD and Quaggin SE (2004) WT1 activates a glomerular-specific enhancer identified from the human nephrin gene. J. Am. Soc. Nephrol 15, 2851–2856 [DOI] [PubMed] [Google Scholar]
- 82.Wagner N, Wagner KD, Xing Y, Scholz H and Schedl A (2004) The major podocyte protein nephrin is transcriptionally activated by the Wilms’ tumor suppressor WT1. J. Am. Soc. Nephrol 15, 3044–3051 [DOI] [PubMed] [Google Scholar]
- 83.Hartkamp J and Roberts SGE (2010) HtrA2, taming the oncogenic activities of WT1. Cell Cycle 9, 2508–2514 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Hartkamp J and Roberts SGE (2008) The role of the Wilms’ tumour-suppressor protein WT1 in apoptosis. Biochem. Soc. Trans 36, 629–631 [DOI] [PubMed] [Google Scholar]
- 85.Mayo MW, Wang CY, Drouin SS, Madrid LV, Marshall AF, Reed JC, Weissman BE and Baldwin AS (1999) WT1 modulates apoptosis by transcriptionally upregulating the bcl-2 proto-oncogene. EMBO J. 18, 3990–4003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Simpson LA, Burwell EA, Thompson KA, Shahnaz S, Chen AR and Loeb DM (2006) The antiapoptotic gene A1/BFL1 is a WT1 target gene that mediates granulocytic differentiation and resistance to chemotherapy. Blood 107, 4695–4702 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Liu XW, Gong LJ, Guo LY, Katagiri Y, Jiang H, Wang ZY, Johnson AC and Guroff G (2001) The Wilms’ tumor gene product WT1 mediates the down-regulation of the rat epidermal growth factor receptor by nerve growth factor in PC12 cells. J. Biol. Chem 276, 5068–5073 [DOI] [PubMed] [Google Scholar]
- 88.Nichols KE, Re GG, Yan YX, Garvin AJ. and Haber DA. (1995) WT1 induces expression of insulin-like growth factor 2 in Wilms’ tumor cells. Cancer Res. 55, 4540–4543 [PubMed] [Google Scholar]
- 89.Scharnhorst V, van der Eb AJ and Jochemsen AG (2001) WT1 proteins: functions in growth and differentiation. Gene 273, 141–161 [DOI] [PubMed] [Google Scholar]
- 90.Maheswaran S, Englert C, Bennett P, Heinrich G and Haber DA (1995) The WT1 gene product stabilizes p53 and inhibits p53-mediated apoptosis. Genes Dev. 9, 2143–2156 [DOI] [PubMed] [Google Scholar]
- 91.Menke AL, Shvarts A, Riteco N, van Ham RCA, van der Eb AJ and Jochemsen AG (1997) Wilms’ tumor 1-KTS isoforms induce p53-independent apoptosis that can be partially rescued by expression of the epidermal growth factor receptor or the insulin receptor. Cancer Res. 57, 1353–1363 [PubMed] [Google Scholar]
- 92.Morrison DJ, English MA and Licht JD (2005) WT1 induces apoptosis through transcriptional regulation of the proapoptotic Bcl-2 family member Bak. Cancer Res. 65, 8174–8182 [DOI] [PubMed] [Google Scholar]
- 93.Szemes M, Dallosso AR, Melegh Z, Curry T, Li Y, Rivers C, Uney J, Mägdefrau AS, Schwiderski K, Park JH et al. (2013) Control of epigenetic states by WT1 via regulation of de novo DNA methyltransferase 3A. Hum. Mol. Genet 22, 74–83 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Kreidberg JA (2010) WT1 and kidney progenitor cells. Organogenesis 6, 61–70 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Morrison AA, Venables JP, Dellaire G and Ladomery MR (2006) The Wilms tumour suppressor protein WT1 (+ KTS isoform) binds α-actinin 1 mRNA via its zinc-finger domain. Biochem. Cell Biol 84, 789–798 [DOI] [PubMed] [Google Scholar]
- 96.Larsson SH, Charlieu JP, Miyagawa K, Engelkamp D, Rassoulzadegan M, Ross A, Cuzin F, van Heyningen V and Hastie ND (1995) Subnuclear localization of WT1 in splicing or transcription factor domains is regulated by alternative splicing. Cell 81, 391–401 [DOI] [PubMed] [Google Scholar]
- 97.Davies RC, Calvio C, Bratt E, Larsson SH, Lamond AI and Hastie ND (1998) WT1 interacts with the splicing factor U2AF65 in an isoform-dependent manner and can be incorporated into spliceosomes. Genes Dev. 12, 3217–3225 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Little NA, Hastie ND and Davies RC (2000) Identification of WTAP, a novel Wilms’ tumour 1-associating protein. Hum. Mol. Genet 9, 2231–2239 [DOI] [PubMed] [Google Scholar]
- 99.Markus MA, Heinrich B, Raitskin O, Adams DJ, Mangs H, Goy C, Ladomery M, Sperling R, Stamm S and Morris BJ (2006) WT1 interacts with the splicing protein RBM4 and regulates its ability to modulate alternative splicing in vivo. Exp. Cell Res 312, 3379–3388 [DOI] [PubMed] [Google Scholar]
- 100.Caricasole A, Duarte A, Larsson SH, Hastie ND, Little M, Holmes G, Todorov I and Ward A (1996) RNA binding by the Wilms’ tumour suppressor zinc finger proteins. Proc. Natl. Acad. Sci. U.S.A 93, 7562–7566 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Ladomery M, Sommerville J, Woolner S, Slight J and Hastie N (2003) Expression in xenopous oocytes shows that WT1 binds transcripts in vivo, with a central role for zinc finger one. J. Cell Sci 116, 1539–1549 [DOI] [PubMed] [Google Scholar]
- 102.Zhai G, Iskandar M, Barilla K and Romaniuk PJ. (2001) Characterization of RNA aptamer binding by the Wilms’ tumor suppressor protein WT1. Biochemistry 40, 2032–2040 [DOI] [PubMed] [Google Scholar]
- 103.Bor YC, Swartz J, Morrison A, Rekosh D, Ladomery M and Hammarskjold ML (2006) The Wilms’ tumor 1 (WT1) gene (+ KTS isoform) functions with a CTE to enhance translation from an unsliced RNA with a retained intron. Genes Dev. 20, 1597–1608 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Dudnakova T, Spraggon L, Slight J and Hastie N (2010) Actin: a novel interaction partner of WT1 influencing its cell dynamic properties. Oncogene 29, 1085–109 [DOI] [PubMed] [Google Scholar]
- 105.Reddy JC, Morris JC, Wang J, English MA, Haber DA, Shi Y and Licht JD (1995) WT1-mediated transcriptional activation is inhibited by dominant negative mutant proteins. J. Biol. Chem 270, 10878–10884 [DOI] [PubMed] [Google Scholar]
- 106.Maheswaran S, Park S, Bernard A, Morris JF, Rauscher FJ III, Hill DE and Haber DA (1993) Physical and functional interaction between WT1 and p53 proteins. Proc. Natl. Acad. Sci. U.S.A 90, 5100–5104 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Idelman G, Glaser T, Roberts CT Jr, and Werner H (2003) WT1–p53 interactions in IGF-I receptor gene regulation. J. Biol. Chem 278, 3474–3482 [DOI] [PubMed] [Google Scholar]
- 108.Scharnhorst V, Dekker P, van der EB AJ and Jochemsen AG (2000) Physical interaction between Wilms tumor 1 and p73 proteins modulates their functions. J. Biol. Chem 275,10202–10211 [DOI] [PubMed] [Google Scholar]
- 109.Discenza M and Pelletier J (2004) Insights into the physiological role of WT1 from studies of genetically modified mice. Physiol. Genomics 16, 287–300 [DOI] [PubMed] [Google Scholar]
- 110.Menke AL, Clarke AR, Leitch A, Ijpenberg A, Williamson KA, Spraggon L, Harrison DJ and Hastie ND (2002) Genetic interactions between the Wilms’ tumor 1 gene and the p53 gene. Cancer Res. 62, 6615–6620 [PubMed] [Google Scholar]
- 111.Stanhope-Baker P, Kessler PM, Li W, Agarwal ML and Williams BR (2004) The Wilms tumor suppressor-1 target gene podocalyxin is transcriptionally repressed by p53. J. Biol. Chem 279, 33575–33585 [DOI] [PubMed] [Google Scholar]
- 112.Busch M, Schwindt H, Brandt A, Beier M, Görldt N, Romaniuk P, Toska E, Roberts S, Royer HD and Royer-Pokora B (2014) Classification of a frameshift/extended and a stop mutation in WT1 as gain of function mutations that activate cell cycle genes and promote Wilms tumour cell proliferation. Hum. Mol. Genet, doi: 10.1093/hmg/ddu111 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Rong Y, Cheng L, Ning H, Zou J, Zhang Y, Xu F, Liu L, Chang Z and Fu XY (2006) Wilms’ tumor 1 and signal transducers and activators of transcription 3 synergistically promote cell proliferation: a possible mechanism in sporadic Wilms’ tumor. Cancer Res. 66, 8049–8057 [DOI] [PubMed] [Google Scholar]
- 114.Maheswaran S, Englert C, Zheng G, Lee SB, Wong J, Harkin DP, Bean J, Ezzell R, Garvin AJ, McCluskey RT et al. (1998) Inhibition of cellular proliferation by the Wilms tumor suppressor WT1 requires association with the inducible chaperone Hsp70. Genes Dev. 12,1108–1120 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Discenza MT, He S, Lee TH, Chu LL, Bolon B, Goodyer P, Eccles M and Pelletier J (2003) WT1 is a modifier of the Pax2 mutant phenotype: cooperation and interaction between WT1 and Pax2. Oncogene 2, 8145–8155 [DOI] [PubMed] [Google Scholar]
- 116.Matsuzawa-Watanabe Y, Inoue J and Semba K (2003) Transcriptional activity of testis-determining factor SRY is modulated by the Wilms’ tumor 1 gene product, WT1. Oncogene 22, 7900–7904 [DOI] [PubMed] [Google Scholar]
- 117.Nachtigal MW, Hirokawa Y, Enyeart-VanHouten DL, Flanagan JN, Hammer GD and Ingraham HA (1998) Wilms’ tumor 1 and Dax-1 modulate the orphan nuclear receptor SF-1 in sex-specific gene expression. Cell 93, 445–454 [DOI] [PubMed] [Google Scholar]
- 118.Wang D, Li Y, Wu C and Liu Y (2011) PINCH1 is transcriptional regulator in podocytes that interacts with WT1 and represses podocalyxin expression. PLoS ONE 6, e17048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Rivera MN, Kim WJ, Wells J, Stone A, Burger A, Coffman EJ, Zhang J and Haber DA (2009) The tumor suppressor WTX shuttles to the nucleus and modulates WT1 activity. Proc. Natl. Acad. Sci. U.S.A 106, 8338–8343 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Rico M, Mukherjee A, Konieczkowski M, Bruggeman LA, Miller RT, Khan S, Schelling JR and Sedor JR (2005) WT1-interacting protein and ZO-1 translocate into podocyte nuclei after puromycin aminonucleoside treatment. Am. J. Physiol. Renal Physiol 289, 431–441 [DOI] [PubMed] [Google Scholar]
- 121.Johnstone RW, See RH, Sells SF, Wang J, Muthukkumar S, Englert C, Haber DA, Licht JD, Sugrue SP, Roberts T et al. (1996) A novel repressor, par-4, modulates transcription and growth suppression functions of the Wilms’ tumor suppressor WT1. Mol. Cell. Biol 16, 6945–6956 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Richard DJ, Schumacher V, Royer-Pokora B and Roberts SGE (2001) Par4 is a coactivator for a splice isoform specific transcriptional activation domain in WT1. Genes Dev. 15,328–339 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Val P, Martinez-Barbera JP and Swain A (2007) Adrenal development is initiated by Cited2 and Wt1 through modulation of Sf-1 dosage. Development 134, 2349–2358 [DOI] [PubMed] [Google Scholar]
- 124.Wang W, Lee SB, Palmer R, Ellisen LW and Haber DA (2001) A functional interaction with CBP contributes to transcriptional activation by the Wilms’ tumor suppressor WT1. J. Biol. Chem 276,16810–16816 [DOI] [PubMed] [Google Scholar]
- 125.Xu B, Zeng DQ, Wu Y, Zheng R, Gu L, Lin X, Hua X and Jin GH (2011) Tumor suppressor menin represses paired box gene 2 expression via Wilms tumor suppressor protein–polycomb group complex. J. Biol. Chem 286, 13937–13944 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Mosevitsky MI (2005) Nerve ending “signal” proteins GAP-43, MARCKS, and BASP1. Int. Rev. Cytol 245, 245–325 [DOI] [PubMed] [Google Scholar]
- 127.Goodfellow SJ, Rebello MR, Toska E, Zeef LA, Rudd SG, Medler KF and Roberts SGE (2011) WT1 and its transcriptional cofactor BASP1 redirect the differentiation pathway of an established blood cell line. Biochem. J 435, 113–125 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Toska ET, Shandilya J, Goodfellow SJ, Medler KF and Roberts SGE (2013) Prohibitin is required for transcriptional repression by the WT1–BASP1 complex. Oncogene, doi: 10.1038/onc.2013.447 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Essafi A, Webb A, Berry RL, Slight J, Burn SF, Spraggon L, Velecela V, Martinez-Estrada OM, Wiltshire JH, Roberts SGE et al. (2011) A Wt1-controlled chromatin switching mechanism underpins tissue-specific wnt4 activation and repression. Dev. Cell 21, 559–574 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Chau YY and Hastie ND (2012) The role of Wt1 in regulating mesenchyme in cancer, development, and tissue homeostasis. Trends Genet. 28, 515–524 [DOI] [PubMed] [Google Scholar]
- 131.Toska E, Campbell HA, Shandilya J, Goodfellow SJ, Shore P, Medler KF and Roberts SGE (2012) Repression of transcription by WT1–BASP1 requires the myristoylation of BASP1 and the PIP2-dependent recruitment of histone deacetylase. Cell Rep. 2, 462–469 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Maekawa S, Murofushi H and Nakamura S (1994) Inhibitory effect of calmodulin on phosphorylation of NAP-22 with protein kinase C. J. Biol. Chem 269, 19462–19465 [PubMed] [Google Scholar]
- 133.Mosevitsky MI, Capony JP, Skladchikova G. Yu., Novitskaya VA, Plekhanov A. Yu. and Zakharov VV (1997) The BASP1 family of myristoylated proteins abundant in axonal termini. Primary structure analysis and physicochemical properties. Biochimie 79, 373–384 [DOI] [PubMed] [Google Scholar]
- 134.Takasaki A, Hayashi N, Matsubara M, Yamauchi E and Taniguchi H (1999) Identification of the calmodulin-binding domain of neuron-specific protein kinase C substrate protein CAP-22/NAP-22. Direct involvement of protein myristoylation in calmodulin-target protein interaction. J. Biol. Chem 274, 11848–11853 [DOI] [PubMed] [Google Scholar]
- 135.Han YQ, Lin Y, Suarez-Saiz F, San-Matina S, Cui J and Minden MD (2008) Wilms’ tumor 1 suppressor gene mediates antiestrogen resistance via down-regulation of estrogen receptor-α expression in breast cancer cells. Mol. Cancer Res 6, 1347–1355 [DOI] [PubMed] [Google Scholar]
- 136.McCarty G, Awad O and Loeb DM (2011) WT1 protein directly regulates expression of vascular endothelial growth factor and is a mediator of tumor response to hypoxia. J. Biol. Chem 286, 43634–43643 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Hanson J, Gorman J, Reese J and Fraizer G (2007) Regulation of vascular endothelial growth factor, VEGF, gene promoter by the tumor suppressor, WT1. Front. Biosci 12, 2279–2290 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Dame C, Kirschner KM, Bartz KV, Wallach T, Hussels CS and Scholz H (2006) Wilms tumor suppressor, Wt1, is a transcriptional activator of the erythropoietin gene. Blood 107, 4282–4290 [DOI] [PubMed] [Google Scholar]
- 139.Reizner N, Maor S, Sarfstein R, Abramovitch S, Welshons WV, Curran EM, Lee AV and Werner H (2005) The WT1 Wilms’ tumor suppressor gene product interacts with estrogen receptor-α and regulates IGF-I receptor gene transcription in breast cancer cells. J. Mol. Endocrinol 35, 135–144 [DOI] [PubMed] [Google Scholar]
- 140.Tajinda K, Carroll J and Roberts CT Jr, (1999) Regulation of insulin-like growth factor I receptor promoter activity by wild-type and mutant versions of the WT1 tumor suppressor. Endocrinology 140, 4713–4724 [DOI] [PubMed] [Google Scholar]
- 141.Werner H, Rauscher FJ III, Sukhatme VP, Drummond IA, Roberts CT Jr, and LeRoith D (1994) Transcriptional repression of the insulin-like growth factor I receptor (IGF-I-R) gene by the tumor suppressor WT1 involves binding to sequences both upstream and downstream of the IGF-I-R gene transcription start site. J. Biol. Chem 269, 12577–12582 [PubMed] [Google Scholar]
- 142.Wagner KD, Wagner N, Schley G, Theres H and Scholz H (2003) The Wilms’ tumor suppressor Wt1 encodes a transcriptional activator of the class IV POU-domain factor Pou4f2 (Brn-3b). Gene 305, 217–223 [DOI] [PubMed] [Google Scholar]
- 143.Wilhelm D and Englert C (2002) The Wilms tumor suppressor WT1 regulates early gonad development by activation of Sf1. Genes Dev. 16,1839–1851 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Loeb DM, Korz D, Katsnelson M, Burwell EA, Friedman AD and Sukumar S (2002) Cyclin E is a target of WT1 transcriptional repression. J. Biol. Chem 277, 19627–19632 [DOI] [PubMed] [Google Scholar]
- 145.Wagner KJ, Patek CE, Miles C, Christie S, Brookes AJ and Hooper ML (2001) Truncation of WT1 results in downregulation of cyclin G1 and IGFBP-4 expression. Biochem. Biophys. Res. Commun 287, 977–982 [DOI] [PubMed] [Google Scholar]
- 146.Lee TH and Pelletier J (2001) Functional characterization of WT1 binding sites within the human vitamin D receptor gene promoter Physiol. Genom 7, 187–200 [DOI] [PubMed] [Google Scholar]
- 147.Duarte A, Caricasole A, Graham CF and Ward A (1998) Wilms’ tumour-suppressor protein isoforms have opposite effects on Igf2 expression in primary embryonic cells, independently of p53 genotype. Br. J. Cancer 77, 253–259 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Madden SL, Cook DM, Morris JF, Gashler A, Sukhatme VP and Rauscher FJ III, (1991) Transcriptional repression mediated by the WT1 Wilms tumor gene product. Science 253, 1550–1553 [DOI] [PubMed] [Google Scholar]
- 149.Ward A, Pooler JA, Miyagawa K, Duarte A, Hastie ND and Caricasole A (1995) Repression of promoters for the mouse insulin-like growth factor II-encoding gene (Igf-2) by products of the Wilms’ tumour suppressor gene Wt1. Gene 167, 239–243 [DOI] [PubMed] [Google Scholar]
- 150.Webster NJ, Kong Y, Sharma P, Haas M, Sukumar S and Seely BL (1997) Differential effects of Wilms’ tumor WT1 splice variants on the insulin receptor promoter. Biochem. Mol. Med 62, 139–150 [DOI] [PubMed] [Google Scholar]
- 151.Werner MH, Huth JR, Gronenborn AM and Clore GM (1996) Molecular determinants of mammalian sex. Trends. Biochem. Sci 21, 302–308 [PubMed] [Google Scholar]
- 152.Wang ZY, Qiu QQ, Enger KT and Deuel TF (1993) A second transcriptionally active DNA-binding site for the Wilms’ tumor gene product, WT1. Proc. Natl. Acad. Sci. U.S.A 90, 8896–8900 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Adachi Y, Matsubara S, Pedraza C, Ozawa M, Tsutsui J, Takamatsu H, Noguchi H, Akiyama T and Muramatsu T (1996) Midkine as a novel target gene for the Wilms’ tumor suppressor gene (WT1). Oncogene 13, 2197–2203 [PubMed] [Google Scholar]
- 154.Hsu SY, Kubo M, Chun SY, Haluska FG, Housman DE and Hsueh AJ (1995) Wilms’ tumor protein WT1 as an ovarian transcription factor: decreases in expression during follicle development and repression of inhibin-alpha gene promoter. Mol. Endocrinol 9, 1356–1366 [DOI] [PubMed] [Google Scholar]
- 155.Wang ZY, Qiu QQ and Deuel TF (1993) The Wilms’ tumor gene product WT1 activates or suppresses transcription through separate functional domains. J. Biol. Chem 268, 9172–9175 [PubMed] [Google Scholar]
- 156.Wang ZY, Qiu QQ, Huang J, Gurrieri M and Deuel TF (1995) Products of alternatively spliced transcripts of the Wilms’ tumor suppressor gene, WT1, have altered DNA binding specificity and regulate transcription in different ways. Oncogene 10, 415–422 [PubMed] [Google Scholar]
- 157.Guan LS, Rauchman M and Wang ZY (1998) Induction of Rb-associated protein (RbAp46) by Wilms’ tumor suppressor WT1 mediates growth inhibition. J. Biol. Chem 273, 27047–27050 [DOI] [PubMed] [Google Scholar]
- 158.Han X and Chesney RW (2003) Regulation of taurine transporter gene (TauT) by WT1. FEBS Lett. 540, 71–76 [DOI] [PubMed] [Google Scholar]
- 159.Hewitt SM, Fraizer GC, Wu YJ, Rauscher FJ and Saunders GF (1996) Differential function of Wilms’ tumor gene WT1 splice isoforms in transcriptional regulation. J. Biol. Chem 271, 8588–8592 [DOI] [PubMed] [Google Scholar]
- 160.Hofmann W, Royer HD, Drechsler M, Schneider S and Royer-Pokora B (1993) Characterization of the transcriptional regulatory region of the human WT1 gene. Oncogene 8, 3123–3132 [PubMed] [Google Scholar]
- 161.Malik KT, Poirier V, Ivins SM and Brown KW (1994) Autoregulation of the human WT1 gene promoter. FEBS. Lett 349, 75–78 [DOI] [PubMed] [Google Scholar]
- 162.Rupprecht HD, Drummond IA, Madden SL, Rauscher FJ and Sukhatme VP (1994) The Wilms’ tumor suppressor gene WT1 is negatively autoregulated. J. Biol. Chem 269, 6198–6206 [PubMed] [Google Scholar]
- 163.Hsu WH, Yu YR, Hsu SH, Yu WC, Chu YH, Chen YJ, Chen CM and You LR (2013) The Wilms’ tumor suppressor Wt1 regulates Coronin 1B expression in the epicardium. Exp. Cell Res 10, 1365–1381 [DOI] [PubMed] [Google Scholar]
- 164.Takeichi M, Nimura K, Mori M, Nakagami H and Kaneda Y (2013) The transcription factors Tbx18 and Wt1 control the epicardial epithelial-mesenchymal transition through bi-directional regulation of Slug in murine primary epicardial cells. PLoS ONE 8, e57829. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Ji SY, Hao JX, Li L, Zhang J, Zheng QS, Li XX, Wang XN, Han CS, Gao F and Liu YX (2013) Expression of inhibin-α is regulated synergistically by Wilms’ tumor gene 1 (Wt1) and steroidogenic factor-1 (Sf1) in sertoli cells. PLoS ONE 8, e53140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Martínez-Estrada OM, Lettice LA, Essafi A, Guadix JA, Slight J, Velecela V, Hall E, Reichmann J, Devenney PS, Hohenstein P et al. (2010) Wt1 is required for cardiovascular progenitor cell formation through transcriptional control of Snail and E-cadherin. Nat. Genet 42, 89–93 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Harrington MA, Konicek B, Song A, Xia XL, Fredericks WJ and Rauscher FJ (1993) Inhibition of colony-stimulating factor-1 promoter activity by the product of the Wilms’ tumor locus. J. Biol. Chem 268, 21271–21275 [PubMed] [Google Scholar]
- 168.Ratelade J, Arrondel C, Hamard G, Garbay S, Harvey S, Biebuyck N, Schulz H, Hastie N, Pontoglio M, Gubler MC et al. (2010) A murine model of Denys–Drash syndrome reveals novel transcriptional targets of WT1 in podocytes. Hum. Mol. Genet 19, 1–15 [DOI] [PubMed] [Google Scholar]
- 169.Andikyan V and Taylor HS (2009) WT1 represses HOX gene expression in the regulation of gynaecologic tumour histologic type. J. Cell. Mol. Med 13, 4522–4531 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Gannon AM and Kinsella BT (2008) The Wilms’ tumour suppressor protein WT1 acts as a key transcriptional repressor of the human thromboxane A2 receptor gene in megakaryocytes. J. Cell. Mol. Med 13, 4571–4586 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Kirschner KM, Hagen P, Hussels CS, Ballmaier M, Scholz H and Dame C (2008) The Wilms’ tumor suppressor Wt1 activates transcription of the erythropoietin receptor in hematopoietic progenitor cells. FASEB J. 22, 2690–2701 [DOI] [PubMed] [Google Scholar]
- 172.Kim J, Prawitt D, Bardeesy N, Torban E, Vicaner C, Goodyer P, Zabel B and Pelletier J (1999) The Wilms’ tumor suppressor gene (Wt1) product regulates Dax-1 gene expression during gonadal differentiation. Mol. Cell Biol 19, 2289–2299 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Englert C, Maheswaran S, Garvin AJ, Kreidberg J and Haber DA (1997) Induction of p21 by the Wilms’ tumor suppressor gene WT1. Cancer Res. 57, 1429–1434 [PubMed] [Google Scholar]
- 174.McCann S, Sullivan J, Guerra J, Arcinas M and Boxer LM (1995) Repression of the c-myb gene by WT1 protein in T and B cell lines. J. Biol. Chem 270, 23785–23789 [DOI] [PubMed] [Google Scholar]
- 175.Kinane TB, Finder JD, Kawashima A, Brown D, Abbate M, Fredericks WJ, Sukhatme VP, Rauscher FJ and Ercolani L (1996) LLC-PK1 cell growth is repressed by WT1 inhibition of G-protein αi-2 protooncogene transcription. J. Biol. Chem 270, 30760–30764 [DOI] [PubMed] [Google Scholar]
- 176.Chen SR, Chen MM, Wang XN, Zhang J, Wen Q, Ji SY, Zheng QS, Gao F and Liu YX. (2013) The Wilms tumor gene, Wt1, mantains testicular cord integrity by regulating the expression of Col4a1 and Col4a2. Biol. Reprod 88, 1–10 [DOI] [PubMed] [Google Scholar]
- 177.Hosono S, Gross I, English MA, Hajra KM, Fearon ER and Licht JD (2000) E-cadherin is a WT1-target gene. J. Biol. Chem 275, 10943–10953 [DOI] [PubMed] [Google Scholar]
- 178.Wu C, Zhu W, Qian J, He S, Wu C, Chen Y and Shu Y (2013) WT1 promotes invasion of NSCLC via suppression of CDH1. J. Thorac. Oncol 9, 1163–1169 [DOI] [PubMed] [Google Scholar]
- 179.Brett A, Pandey S and Fraizer G (2013) The Wilms’ tumor gene (WT1) regulates E-cadherin expression and migration of prostate cancer cells. Mol. Cancer 12, 3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Kirschner KM, Wagner N, Wagner KD, Wellmann S and Scholz H (2006) The Wilms’ tumor suppressor Wt1 promotes cell adhesion through transcriptional activation of the α4integrin gene. J. Biol. Chem 281, 31930–31939 [DOI] [PubMed] [Google Scholar]
- 181.Wagner N, Wagner KD, Scholz H, Kirschner KM and Schedl A (2006) Intermediate filament protein nestin is expressed in developing kidney and heart and might be regulated by the Wilms’ tumor suppressor Wt1. Am. J. Physiol. Regul. Integr. Comp. Physiol 291, 779–787 [DOI] [PubMed] [Google Scholar]
- 182.Jacobi CL, Rudigier LJ, Scholz H and Kirschner KM (2013) Transcriptional regulation by the Wilms’ tumor protein, Wt1, suggests a role of the metalloproteinase Adamts16 in murine genitourinary development. J. Biol. Chem 26, 18811–18824 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Morrison DJ, Kim MK, Berkofsky-Fessler W and Licht JD (2008) WT1 induction of MAP kinase phosphatase 3 represents a novel mechanism of growth suppression. Mol. Cancer Res 6, 1225–1231 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Han Y, San-Marina S, Liu J and Minden MD (2004) Transcriptional activation of c-myc proto-oncogene by WT1 protein. Oncogene 23, 6933–6941 [DOI] [PubMed] [Google Scholar]
- 185.Udtha M, Lee SJ, Alam R, Coombes K and Huff V (2003) Upregulation of c-MYC in WT1-mutant tumors: assessment of WT1 putative transcriptional targets using cDNA microarray expression profiling of genetically defined Wilms’ tumors. Oncogene 22, 3821–3826 [DOI] [PubMed] [Google Scholar]
- 186.Cheema SK, Mishra SK, Rangnekar VM, Tari AM, Kumar R and Lopez-Berestein G (2003) Par-4 transcriptionally regulates Bcl-2 through a WT1-binding site on the bcl-2 promoter. J. Biol. Chem 278, 19995–20005 [DOI] [PubMed] [Google Scholar]
- 187.Zhang X, Xing G and Saunders GF (1999) Proto-oncogene N-myc promoter is down regulated by the Wilms’ tumor suppressor gene WT1. Anticancer Res. 19, 1641–1648 [PubMed] [Google Scholar]
- 188.Ritchie MF, Yue C, Zhou Y, Houghton PJ and Soboloff J (2010) Wilms tumor suppressor 1 and early growth response 1 are regulators of STIM1 expression. J. Biol. Chem 285, 10591–10596 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Steege A, Fahling M, Martinka P, Patzak A, Brinkmeier T, Persson PB, Scholz H and Mrowka R (2008) The Wilms tumor suppressor protein (WT1) splice variants WT1 (−KTS) and WT1 (+KTS) show distinct influences on different control levels of the Renin gene expression. FASEB J. 22, 969.16 [Google Scholar]
- 190.Johannesen J, Karlsen AE, Pociot F, Roenn SG and Nerup J (2003) Strain dependent rat iNOS promoter activity correlation to identified WT1 transcription factor binding site. Autoimmunity 36, 167–175 [DOI] [PubMed] [Google Scholar]
- 191.Marcet-Palacios M, Marcet-Palacios M, Ulanova M, Duta F, Puttagunta L, Munoz S, Gibbings D, Radomski M, Cameron L, Mayers I and Befus AD (2007) The transcription factor Wilms tumor 1 regulates matrix metalloproteinase-9 through a nitric oxide-mediated pathway. J. Immunol 179, 256–265 [DOI] [PubMed] [Google Scholar]
- 192.Oh S, Song Y-H, Kim U-J, Yim J and Kim TK (1999) In vivo and in vitro analyses of c-Myc for differential promoter activities of human telomerase (hTERT) gene in normal and tumor cells. Biochem. Biophys. Res. Commun 263, 361–365 [DOI] [PubMed] [Google Scholar]
- 193.McCoy C, McGee SB and Cornwell MM (1999) The Wilms’ tumor suppressor, WT1, inhibits 12-O-tetradecanoylphorbol-13-acetate activation of the multidrug resistance-1 promoter. Cell Growth Differ. 10, 377–386 [PubMed] [Google Scholar]
- 194.Dejong V, Degeorges A, Filleur S, Ait-Si-Ali S, Mettouchi A, Bornstein P, Binetruy B and Cabon F (1999) The Wilms’ tumor gene product represses the transcription of thrombospondin 1 in response to overexpression of c-Jun. Oncogene 18, 3143–3151 [DOI] [PubMed] [Google Scholar]
- 195.Moshier JA, Skunca M, Wu W, Boppana SM, Rauscher FJ III and Dosescu J (1996) Regulation of ornithine decarboxylase gene expression by the Wilms’ tumor suppressor WT1. Nucleic Acids Res. 24, 1149–1157 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Bourkoula K, Englert C, Giaisi M, Kohler R, Krammer PH and Li-Weber M (2013) The Wilms’ tumor suppressor WT1 enhances CD95L expression and promotes activation-induced cell death in leukemic T cells. Int. J. Cancer 134, 291–300 [DOI] [PubMed] [Google Scholar]
- 197.Lee TH, Lwu S, Kim J and Pelletier J (2002) Inhibition of Wilms tumor 1 transactivation by bone marrow zinc finger 2, a novel transcriptional repressor. J. Biol. Chem 277, 44826–44837 [DOI] [PubMed] [Google Scholar]
- 198.Florio F, Cesaro E, Montano G, Izzo P, Miles C and Costanzo P (2010) Biochemical and functional interaction between ZNF244 and ZNF255, two members of the Krüppel-like zinc-finger protein family and WT1 protein isoforms. Hum. Mol. Genet 19, 3544–3556 [DOI] [PubMed] [Google Scholar]
- 199.Montano G, Cesaro E, Fattore L, Vidovic K, Palladino C, Crescitelli R, Izzo P, Turco MC and Costanzo P (2013) Role of WT1–ZNF224 interaction in the expression of apoptosis-regulating genes. Hum. Mol. Genet 9, 1771–1782 [DOI] [PubMed] [Google Scholar]
- 200.Srichai MB, Konieczkowski M, Padiyar A, Konieczkowski DJ, Mukherjee A, Hayden PS, Kamat S, El-Meanawy MA, Khan S, Mundel P et al. (2004) A WT1 co-regulator controls podocyte phenotype by shuttling between adhesion structures and nucleus. J. Biol. Chem 279, 14398–14408 [DOI] [PubMed] [Google Scholar]
- 201.Du X, Hublitz P, Gunther T, Wilhelm D, Englert C and Schule R (2002) The LIM-only coactivator FHL2 modulates WT1 transcriptional activity during gonadal differentiation. Biochim. Biophys. Acta 1577, 93–101 [DOI] [PubMed] [Google Scholar]
- 202.Johnstone RW, Wang J, Tommerup N, Vissing H, Roberts T and Shi Y (1998) Ciao 1 is a novel WD40 protein that interacts with the tumor suppressor protein WT1. J. Biol. Chem 273,10880–10887 [DOI] [PubMed] [Google Scholar]
- 203.Spraggon L, Dudnakova T, Slight J, Lustig-Yariv O, Cotterell J, Hastie N and Miles C (2007) hnRNP-U directly interacts with WT1 and modulates WT1 transcriptional activation. Oncogene 26, 1484–1491 [DOI] [PubMed] [Google Scholar]
- 204.Maheswaran S, Englert C, Lee SB, Ezzel RM, Settleman J and Haber DA. (1998) E1B 55K sequesters WT1 along with p53 within a cytoplasmic body in adenovirus-transformed kidney cells. Oncogene 16, 2041–2050 [DOI] [PubMed] [Google Scholar]
- 205.Kim JM, Hong Y, Semba K and Kim S (2000) Physical and functional interaction between HCMV IE2 protein and the Wilms’ tumor suppressor WT1. Biochem. Biophys. Res. Commun 267, 59–63 [DOI] [PubMed] [Google Scholar]