Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Feb 17.
Published in final edited form as: Biosci Rep. 2012 Apr 1;32(2):113–130. doi: 10.1042/BSR20110046

FOXA1: a transcription factor with parallel functions in development and cancer

Gina M BERNARDO *, Ruth A KERI *,†,‡,1
PMCID: PMC7025859  NIHMSID: NIHMS590780  PMID: 22115363

Synopsis

When aberrant, factors critical for organ morphogenesis are also commonly involved in disease progression. FOXA1 (forkhead box A1), also known as HNF3α (hepatocyte nuclear factor 3α), is required for postnatal survival due to its essential role in controlling pancreatic and renal function. In addition to regulating a variety of tissues during embryogenesis and early life, rescue experiments have revealed a specific role for FOXA1 in the postnatal development of the mammary gland and prostate. Activity of the nuclear hormone receptors ERα (oestrogen receptor α) and AR (androgen receptor) is also required for proper development of the mammary gland and prostate respectively. FOXA1 modulates ER and AR function in breast and prostate cancer cells, supporting the postulate that FOXA1 is involved in ER and AR signalling under normal conditions, and that some carcinogenic processes in these tissues stem from hormonally regulated developmental pathways gone awry. In addition to broadly reviewing the function of FOXA1 in various aspects of development and cancer, this review focuses on the interplay of FOXA1/ER and FOXA1/AR, in normal and cancerous mammary and prostate epithelial cells. Given the hormone dependency of both breast and prostate cancer, a thorough understanding of FOXA1’s role in both cancer types is critical for battling hormone receptor-positive disease and acquired anti-hormone resistance.

Keywords: androgen receptor (AR), breast, foxhead box A1 (FOXA1), foxhead box A2 (FOXA2), oestrogen receptor (ER), prostate

INTRODUCTION

FOXA1 (forkhead box A1) is the founding member of the FOX family of transcription factors that is comprised of at least 40 members (reviewed in [1]). FOXA1/HNF3α (hepatocyte nuclear factor 3α), FOXA2/HNF3β and FOXA3/HNF3γ constitute the FOXA subfamily, which was originally identified for its transcriptional regulation of the genes liver-specific (Ttr) transthyretin and α1-antitrypsin (Serpinal) [2]. Since this seminal study, FOXAs have been found to regulate many genes involved in developmental specification of not just hepatic, but several other tissues (reviewed in [3]). FOXAs contain an ~ 100 amino acid DNA-binding domain or FOX/winged helix domain that is highly conserved (at least 92%) within the FOXA family [4], and shares extremely high homology (90%) with that of its namesake, the Drosophila homologue fkh (forkhead) [4,5]. FOXAs also contain conserved nuclear localization sequences and homology in the N- and C-terminal transactivation domains [4,6,7]. FOXAs bind as monomers [8] to the consensus element A(A/T)TRTT(G/T)RYTY [9] and crystallization of the DNA-binding domain of FOXA3 revealed a ‘winged helix’ structure bound to DNA in a manner similar to that of linker histones [8]. Importantly, unlike linker histones, FOXAs lack the basic amino acids required for chromatin compaction [10]. Thus, FOXA binding to nucleosomes induces an open chromatin configuration enabling the recruitment of other transcriptional regulators[1012]. This function has led to FOXAs being coined as ‘pioneering’ or ‘licensing’ factors. Recently, the participation of FOXA1 in chromatin remodelling has been further described, where FOXA1 binding to DNA precedes the loss of cytosine methylation and the dimethylation of histone H3 lysine K4 (H3K4) during the differentiation of pluripotent P19 cells [13].

The ability of FOXAs to remodel heterochromatin provides a mechanistic basis for how these factors initiate transcriptional cascades involved in both development and disease. Specifically, FOXA1 is required for normal development of the mammary gland and prostate, and is necessary for both ER (oestrogen receptor)- and AR (androgen receptor)-regulated transcription in hormone receptor-positive breast and prostate cancers respectively. An understanding of FOXA1 during development has generated great insight into its function in cancer progression and vice versa. This review focuses on the similarities and differences between the normal developmental role of FOXA1 and that involved in cancer.

FOXA1 IN DEVELOPMENT

Identifying FOXAs as transcriptional regulators of hepatic specification [2] led multiple groups to perform expression pattern analyses for these factors from early development throughout adulthood. Foxa2 mRNA is the first to be expressed during embryogenesis, and is observed during gastrulation in the anterior primitive streak and node with subsequent expression in the notochord, floor plate and gut [1416]. Foxa1 becomes detectable at the late primitive streak stage in the midline endoderm of mouse embryos, followed by expression in the ventral floorplate, notochord and gut [1416]. Foxa3 is the last to be activated, being expressed during hindgut differentiation [15]. In the adult mouse, expression of FOXA1 and FOXA2 is observed within endoderm-, mesoderm- and ectoderm-derived tissues [17,18]. Foxa3 mRNA is less restricted in adult tissues, being present within the heart, adipose tissue, ovary and testis, in addition to endoderm-derived liver and gastrointestinal tissues [17]. These results, in combination with the chromatin-remodelling function observed for FOXA, provided evidence that FOXAs may function in developmental specification. Through both germ-line and conditional knockout approaches, the functions of FOXA1, FOXA2 and FOXA3 have been investigated both independently and, in the case of FOXA1 and FOXA2, in combination, and each has been proven to be required during various aspects of development (reviewed in [1,3,19]). Briefly, the loss of Foxa2 is embryonic lethal due to failed node and notochord development [20]. Foxa1 null mice survive through embryogenesis, but are postnatally lethal due to severe hypoglycaemia and dehydration as described in detail below [21,22]. In contrast, Foxa3 knockout mice are normal, developing without morphological defects through adulthood [23], but are hypoglycaemic in response to fasting [24] and the males are subfertile [25]. The severity of these respective phenotypes parallels the onset of expression of Foxa2, then Foxa1 and then Foxa3 during embryogenesis, and suggests compensatory roles for FOXA2 in the Foxa1 knockout mice, and for both FOXA2 and FOXA1 in Foxa3 knockout mice. The involvement of FOXA1, specifically, will be discussed herein and is overviewed in Table 1.

Table 1.

Overview of Foxa1 mouse models of development

Tissue Origin Genotype Viability Phenotype Foxa2 redundancy Reference(s)
Pancreas Endoderm Foxa1−/− Lethal, P2–12/14 Insulin secretion defect; hypoglycaemia; hypotriglyceridaemia No [21,22,26]
Pdx1-CreE/FoxaloxP/loxP Adulthood No major defects Yes [27]
Pdx1-CreE/Foxa1loxP/loxP/Foxa2loxP/+ Adulthood No major defects Yes [27]
Pdx1-CreE/Foxa1loxP/+ /Foxa2loxP/loxP Lethal, P5 Hypoplasia n/a [27]
Pdx1-CreE/Foxa1loxP/loxP/Foxa2loxP/loxP Lethal, P2 Failed endocrine and exocrine differentiation; hypoplasia n/a [27]
Kidney Mesoderm Foxa1−/− Lethal, P2–12 Dehydration; nephrogenic diabetes insipidus No [21,30]
Liver Endoderm Foxa1−/− /Foxa3Cre/Foxa2loxP/loxP Lethal, E10 Failed fetal liver development n/a [31]
Alf-pCre/Foxa1loxP/loxP/Foxa2loxP/loxP Adulthood Bile duct hyperplasia; fibrosis n/a [32]
Lung Endoderm Foxa1−/− Lethal, P2–13 Delayed alveolarization; surfactant secretion defect Yes [34]
Foxa1−/− /SPC-rtTA−/tg/(tetO)7Cre−/tg/Foxa2loxP/loxP At least E18.5 Impaired branching morphogenesis
Brain Ectoderm Foxa1−/− Lethal, P2–12 Delayed dopaminergic neuron maturation Yes [44]
Foxa1−/− /Nestin-Cre/Foxa2loxP/loxP* At least E18.5 Failed dopaminergic neuron maturation n/a [44]
En1-klCre/Foxa1loxP/loxP/Foxa2loxP/loxP At least E11.5 Failed dopaminergic neuron maturation n/a [48]
Foxa1−/− /Wnt1-Cre/Foxa2loxP/loxP At least E18.5 Failed ventral midbrain progenitor specification n/a [49]
GI tract Endoderm Villin-Cre/Foxa1loxP/loxP/Foxa2loxP/loxP Adulthood Impaired goblet and enterendocrine cell maturation; mucin secretion defect n/a [50]
Prostate Endoderm Foxa1−/− Rescued Failed luminal lineage differentiation; epithelial hyperproliferation; secretion defect No [58]
Mammary Ectoderm Foxa1−/− Rescued Impaired ductal invasion No [73]
*

Investigated the dose-dependent effect of each allele of FOXA1/A2. For simplicity, these combinations were not reported here. n/a, not applicable; in these models Foxa2 was deleted in combination with Foxa1. GI, gastrointestinal.

Pancreas and kidney

Mice that are homozygous null for Foxa1 lack any overt morphological abnormalities [21,22]. However, they are postnatally lethal due to a combined phenotype of severe hypoglycaemia and dehydration. Interestingly, loss of Foxa1 also dramatically decreases circulating glucagon levels. This is paradoxical to the observed hypoglycaemic state because the pancreas normally responds to hypoglycaemia by releasing glucagon, which then induces conversion of glycogen stores into glucose in the liver. Further investigation revealed that FOXA1 is normally expressed in glucagon-expressing pancreatic α-islet cells, where it is necessary to transcriptionally activate the proglucagon promoter (Gcg) [21]. Loss of Foxa1 reduces glucagon transcription, subsequently decreasing glucagon secretion into the circulation. These results confirm a role for FOXA1 in regulating glucose homoeostasis through directing α-islet cell function. In addition to the loss of proglucagon gene expression, Foxa1 null mice also fail to secrete insulin in response to glucose administration [22]. This defect is attributed to the up-regulation of mitochondrial Ucp2 (uncoupling protein 2) that occurs in the pancreatic β-cells of Foxal null mice [26]. Increased UCP2 expression uncouples oxidative phosphorylation, which decreases glucose-mediated ATP synthesis and insulin secretion from β-islet cells. Together, these studies revealed that FOXA1 modulates glucose homoeostasis through multiple mechanisms: transcriptional activation of the proglucagon gene and repression of Ucp2 expression.

To investigate the role of FOXA1 during pancreatic development without the physiological complexity of a global knockout, mice were generated with loxP sites flanking exon 2 of Foxa1. These mice were then crossed with Pdx1-CreEarly transgenics mice [27]. PDX-1 (pancreas and duodenal homeobox protein-1) expressing cells generate each cell type (exocrine, endocrine and duct) of the pancreas allowing for pancreas-selective expression of Cre DNA recombinase and disruption of floxed transgenes [28]. Surprisingly, Pdx1-CreEarly; Foxa1loxP/loxP mice are viable and fertile indicating that the postnatal lethality observed in the germ-line knockouts [21] was not solely due to an isolated defect in the pancreas, but likely also involved severe dehydration [27]. Compound conditional knockout mouse studies subsequently revealed that FOXA1 and FOXA2 co-operatively control pancreatic acinar and islet morphogenesis [27]. The presence of at least one wild-type allele of Foxa2 can compensate for complete loss of Foxa1 in the pancreas, resulting in viable mice. Interestingly, the absence of one or two wild-type alleles of Foxa1 dictates the level of specification of exocrine and endocrine cell lineages in the pancreas of mice completely lacking Foxa2 [27,29]. In other words, mice that are wild-type, heterozygous null and homozygous null for Foxa1 in the pancreas have the most normal, intermediary and severe disruption respectively of pancreatic lineage specification when combined with the homozygous null Foxa2 allele. For a detailed description of FOXA1/2 compound mutants that have been generated to investigate their functions in the pancreas and other tissues, see [3,19]. Notably, mice null for Foxa3 are hypoglycaemic when fasted due to decreased expression of the GLUT2 (glucose transporter) [24], and thus each FOXA family member is necessary for maintaining glucose homoeostasis.

In addition to the deregulation of pancreatic proglucagon and Ucp2 expression, pups lacking FOXA1 are severely dehydrated [21], prompting studies to investigate FOXA1 function in the kidney. Foxa1 null mice develop nephrogenic diabetes insipidus as shown by the inability to respond to arginine-vasopressin [ADH (anti-diuretic hormone)] [30]. Interestingly, the development of this disease could not be explained by a decrease in the expression of genes encoding the vasopressin 2 receptor, aquaporins or other proteins involved in water reabsorption. Although the precise mechanism by which FOXA1 modulates kidney function remains to be determined, it has been postulated that the lethality of the Foxa1 null animals occurs as a result of the combined impact of pancreatic insufficiency and kidney defects. This is supported by the viability of mice with a pancreatic specific deletion of Foxa1 (Pdx1-CreEarly; Foxa1loxP/oxP) as discussed above. It is unclear whether the kidney defect in Foxa1 null mice is due to a direct effect on this organ or changes that occur in response to an abnormal developmental programme. Use of a kidney-specific inducible knockout would facilitate addressing this question, as well as provide a resource for identifying the specific genes which are controlled by FOXA1 that ultimately establish arginine-vasopressin responsiveness. In addition, such animals would generate a potential model for nephrogenic diabetes insipidus in humans. Identifying the specific role FOXA1 plays in water reuptake should ultimately provide important insights for clinical management of this disease.

Liver

Given the role for FOXA proteins in regulating liver-specific gene transcription [2], it was hypothesized that FOXA1 would be necessary for normal liver development. However, while Foxa1 null mice die postnatally [21], they have a morphologically normal liver. Similar to the maintenance of normal pancreatic morphology in Foxa1 knockouts due to FOXA2 compensation, FOXA2 also offsets loss of Foxa1 in the liver. When investigated combinatorially, genomic loss of Foxa1, within the context of an endoderm-specific conditional deletion of Foxa2 (Foxa3-Cre; Foxa2loxP/loxP), completely blocked the onset of liver specification whereas conditional loss of Foxa2 alone had no effect [31]. The endodermal disruption of Foxa2 occurs at E8.5 (embryonic day 8.5) when using Foxa3-Cre to induce recombination, and this precedes liver specification. To investigate liver development after the onset of specification, Alf-pCre; Foxa1loxp/loxp; Foxa2loxp/loxp mice were generated [32]. In these mice, Cre expression occurs at E10.5 in the liver primordium, but complete loss of Foxa1/2 was not observed until P2 (postnatal day 2). These mice undergo normal hepatocyte differentiation, but during adulthood develop bile duct hyperplasia and fibrosis as a result of increased IL-6 (interleukin-6) expression. Under normal conditions, FOXA1/2 and the GR (glucocorticoid receptor) co-operate to repress Il6 transcription. In the absence of FOXA1/2, this repression is ablated, allowing NF-κB (nuclear factor κB)-induced up-regulation of Il6. The excessive IL-6 then induces proliferation of bile duct epithelial cells (cholangiocytes) [32]. These results demonstrate the combined importance of FOXA1 and FOXA2 in liver cell specification and differentiation, and suggest a potential role for FOXA1/2 in human diseases of the liver. Along these lines, FOXA2 expression is decreased in the livers of humans with cholestatic disease [33], but a role for FOXA1 in biliary diseases remains to be established.

Lung

FOXA1 is observed in the lung bud at E10.5, and is maintained in mature conducting airway epithelial and secretory alveolar type II cells [18]. Mice null for Foxa1 have delayed lung development [34]. Normally, the fetal lung develops through three distinct morphological phases: pseudoglandular, canalicular and saccular. While lungs of wild-type mice reach the saccular stage by E16.5, those lacking FOXA1 are delayed in the canalicular stage until E17.5. Similarly, lung septation in postnatal Foxa1 knockout mice (P5) was significantly delayed. Loss of Foxa1 also decreased the expression of lung differentiation markers [e.g. CCSP (clara cell secretory protein), SP-B (pro-surfactant protein-B) and SP-C (pro-surfactant protein-C)] during embryogenesis. This is at least partly due to the dependency of both CCSP (Scbglal) [35] and SP-B (Sftpb) [36,37] promoters on FOXA1 for activation in lung epithelium. How FOXA1 modulates SP-C expression is not known. Although FOXA1 deficiency causes a developmental delay during embryogenesis, histology of the knockout lungs was indistinguishable from controls by P13. That said, cuboidal type II cells had higher glycogen levels, a decreased number of lamellar bodies and less surfactant than those in wild-type lungs. As in other tissues (e.g. pancreas and liver), FOXA2 eventually compensates for loss of Foxa1 in the lung. Complete loss of Foxa1, combined with doxycycline-induced deletion of Foxa2 driven by Cre expression under control of the SP-C lung-specific promoter [Foxa1−/− /SPC-rtTAltg/(tetO)7Creltg/Foxa2loxP/loxP] during embryogenesis, drastically impairs lung branching morphogenesis and epithelial differentiation [38]. Similar to Foxa1 null lungs, CCSP expression is absent in the compound mutants. In addition, loss of Foxa1 and Foxa2 decreases mRNA expression of sonic hedgehog (Shh), a necessary activator of lung epithelial specification [39,40]. FOXA1 expression is increased in a rat model of ALI (acute lung injury), in which it is suggested to function in alveolar type II epithelial cell apoptosis [41], and is similarly necessary for H2O2-induced apoptosis in A549 cells [42]. Together, these results imply FOXA1 may participate in the onset or progression of lung diseases not limited to cancer as discussed below. It will be important to generate mice with lung specific conditional deletion or transgenic overexpression of FOXA1 to gain mechanistic insight into the precise role FOXA1 may play in these diseases.

Brain

FOXA1 and FOXA2 are expressed in the notochord and floor-plate of the fetal brain [1416], and Foxa2 null mice are embryonic lethal as a result of failed node and notochord development [20,43]. Although Foxa1 null pups do not exhibit overt CNS (central nervous system) architectural defects [21,22], they have delayed maturation of mature mDA (midbrain dopaminergic) neurons [44]. Conditional loss of Foxa2 using a Nestin-Cre transgene dose-dependently exacerbates the Foxa1 null phenotype, indicating that even one copy of Foxa2 compensates for Foxa1 loss and vice versa. The phenotypic response to FOXA loss has been attributed to the ability of these proteins to control expression of neurogenin 2, Nurr1 and EN1 (engrailed 1), three factors essential for neuronal differentiation [4547]. Complimenting these studies, conditional loss of both Foxal and Foxa2, through breeding with En1-klCre transgenic mice, led to disruption of each FOXA protein earlier in development (E9.75) [48] compared with Nestin-Cre (E10.5)-mediated disruption [44]. Through these two approaches, Lin et al. [48] showed that FOXA1/2 positively regulate expression of the LIM homeodomain transcription factors Lmx1a/1b and negatively regulate the morphogenically associated homeodomain protein Nkx2.2 earlier in development. Intriguingly, FOXA1 and FOXA2 are redundantly necessary for the initiation of SHH expression in the midbrain [48,49], paralleling the regulation of Shh by FOXA1 observed in the lung that was discussed previously. Further demonstrating the importance of FOXA in SHH signalling, at least in this context, FOXA1/2 are also responsible for restricting the expression of Gli1 and Gli2, transcriptional mediators of SHH signalling, and patched-1, the receptor for SHH, in the ventral midbrain [49].

Gastrointestinal tract

FOXA1/2 are also expressed in the epithelia of the gastrointestinal tract [18]. To investigate how the combined loss of Foxa1 and Foxa2 specifically affects intestinal development, floxed alleles were conditionally disrupted using the villin promoter to direct expression of Cre (Vil-Cre) to the intestinal epithelium [50]. The compound-conditional mutants are viable, but grow slower, and have decreased body weight due to a reduction in lean muscle mass and less body fat. Although overall intestinal morphology is normal, the number, secretory capacity and differentiation of goblet cells are reduced in the combined mutants. FOXA1, but not FOXA2, was found to bind to the promoter of the Muc2 gene that encodes mucin 2. These results compliment previous in vitro studies in which FOXA1 and FOXA2 can both transactivate the Muc2 promoter, with FOXA1 being more efficient [51]. In addition to diminished goblet cell formation, loss of Foxa1/2 leads to decreased differentiation of L- and D-cell enteroendocrine lineages [51], which produce hormones involved in intestinal function. Paralleling in vitro regulation of the proglucagon promoter by FOXA1/2 [21,52], intestines of the combined conditional mutants lack D-cells, as shown by loss of cells expressing GLP (glucagon-like peptides)-1 and -2. In addition, cell populations that express somatostatin and PYY (peptide YY) (D- and L-cells respectively) were decreased. Furthermore, expression of Islet-1 and Pax6, two transcription factors involved in enteroendocrine cell differentiation, are decreased, providing evidence that FOXA1/2 precede Islet-1 and Pax6 expression in the enteroendocrine transcriptional hierarchy. These results indicate that FOXA proteins are essential for development of both mucin-producing goblet and enteroendocrine cellular populations. The loss of these cell types probably results in insufficient nutrient absorption required for normal growth, thus providing an explanation for the decreased growth rate of these mice during early life.

Prostate

Morphogenesis of the prostate is dependent on stromal AR expression, whereas prostatic secretory function relies on epithelial AR expression [53,54]. FOXA1 is an established regulator of AR transcriptional activity in prostate cancer cells (reviewed below) leading to an investigation of FOXA1 during prostate development. Expression analyses revealed that FOXA1 is present in prostate tissue from the onset of development at E18 in the urogenital sinus epithelium throughout adulthood, with no detectable expression in the stroma [5557]. Considering the postnatal lethality of the Foxa1 null mice, Matusik and co-workers [58] investigated prostate morphogenesis in the absence of FOXA1 through utilization of two distinct rescue strategies: renal capsule grafting and tissue recombination. While loss of Foxa1 does not alter rudiment formation, luminal epithelial lineage differentiation is blocked as shown by luminal CK8 (cytokeratin 8) staining being present only in concert with the basal epithelial lineage marker p63. Furthermore, CK5-positive basal cell and α-SMA (α-smooth muscle actin)-positive smooth muscle cell populations were aberrantly expanded. The epithelium within the Foxa1 null prostate also fails to polarize and undergo proper lumen formation, and, as expected, results in a secretion defect. While prostatic tissue lacking epithelial AR has a similar secretory phenotype [54], Foxa1 null prostates maintain epithelial AR expression. Since FOXA1 facilitates AR transcriptional activity as discussed below, it is possible that while FOXA1 is not required for AR expression, it is necessary for AR function in the developing prostate, thus resulting in the observed secretory defect. This is supported by the loss of Pbsn (probasin) and Sbp (spermine-binding protein) mRNA, which are normally stimulated by AR activation, in the Foxa1 null prostate glands [58].

Interestingly, FOXA2 is aberrantly up-regulated in Foxa1 knockout prostate epithelium [58]. Normally, FOXA2 is only found in prostatic epithelia during embryogenesis, and in a small population of mature basal epithelial cells that co-express syn-aptophysin, an NE (neuroendocrine) marker [57,59]. Thus, unlike in other tissues (e.g. lung, liver and pancreas), FOXA2 does not compensate for FOXA1 loss in the prostate. As discussed below with regard to prostate cancer, the up-regulation of FOXA2 in the Foxa1 null prostates may enable AR function in an androgen-independent manner, resulting in the observed hyper-proliferative phenotype. Of note, loss of Foxa1 also decreases expression of the homeobox gene Nkx3.1, a putative tumour suppressor whose deficiency also leads to prostatic hyperplasia [60]. Prostates lacking FOXA1 also have increased SHH, an established contributor to prostate carcinogenesis [6163]. Interestingly, the negative regulation of SHH by FOXA1 in the prostate differs from the positive regulation observed in lung and brain. This may be due to the absence of FOXA1/FOXA2 collaboration in the prostate, but this possibility has not yet been tested.

Mammary gland

In breast cancer, FOXA1 expression positively correlates with that of ER and another transcription factor, GATA3 [6466], both of which are necessary mediators of normal mammary gland development [6770]. This strong correlation, the known up-regulation of FOXA1 as a result of GATA3 overexpression in HEK-293T cells [HEK-293 cells expressing the large T-antigen of SV40 (simian virus 40)] [71], and the co-expression of FOXA1 and GATA3 in mouse mammary epithelia, prompted Kouros-Mehr et al. [70] to investigate whether GATA3 could regulate FOXA1 expression in the normal mammary gland. Using ChIP (chromatin immunoprecipitation), the authors showed that GATA3 binds to the Foxal promoter in mouse primary mammary epithelial cells. Further investigation into a mammary-specific role for the GATA3-interacting protein, FOG2 (friend of GATA2), revealed that conditional disruption of Fog2 by WAP (whey acidic protein)-Cre-mediated excision resulted in decreased Foxal mRNA expression during involution [72]. However, limitations in the technical design of this study, specifically the lack of epithelial-specific normalization approaches, leave open the possibility that FOG2 does not directly regulate Foxal expression. A more recent study investigating FOXA1 protein expression in virgin adult mammary glands lacking Gata3 following MMTV (murine mammary tumour virus)-Cre-induced conditional deletion failed to detect a change in the percentage of FOXA1-positive cells in response to Gata3 disruption compared with wild-type controls [73]. These results indicate that GATA3 is not required for FOXA1 expression in the normal mammary gland. However, they do not negate GATA3 binding to the Foxal promoter in primary mammary cells [70] and leave open the possibility that GATA3 may regulate FOXA1 expression in other tissue types [71].

The observed correlation between FOXA1 and ER in human breast cancer also led to in vitro functional studies that revealed an essential role for FOXA1 in mediating transcriptional regulation by ER, as reviewed below. These results suggested that FOXA1 may be necessary during mammary morphogenesis simply due to its requirement for full ER activity. Supporting this hypothesis, FOXA1 and ER are co-expressed in mammary luminal epithelium during development [73]. To directly evaluate the role for FOXA1 in this process, we rescued Foxa1 null mammary tissue via renal capsule grafting and orthotopic transplantation [73]. Loss of Foxa1 completely blocks epithelial outgrowth in the orthotopic transplant model, and inhibits ductal invasion in renally transplanted glands. Unlike in the prostate, Foxal knockout mammary epithelium exhibits no observable alterations in the lineage markers α-SMA, CK8 and E-cadherin. Another difference between mammary and prostate responses to FOXA1 loss involves expression of their cognate steroid hormone receptors. While AR expression is maintained in the Foxal null prostate, loss of Foxal in the mammary gland is accompanied by a lack of epithelial ER expression. In support of these in vivo findings, FOXA1 binds to the ESRl promoter and is required for ER mRNA and protein expression in breast cancer cells [73]. Combined, these results revealed that FOXA1 is not only required for ER activity but also for its expression. The transcriptional hierarchy of FOXA1, ER and GATA3 has also been further refined. Mammary glands deficient in ER maintain expression of FOXA1 and Gata3 [73]. In contrast, mammary glands deficient in GATA3 maintain FOXA1 [73], but lose ER [69,70]. Together, these results place both GATA-3 and FOXA1 upstream of ER, and also reveal that the expression of GATA-3 and FOXA1 are not interdependent. Interestingly, unlike the secretary defects seen in the pancreas, lung, intestine and prostate discussed above, loss of Foxal in the mammary gland does not affect lobulo-alveolar maturation and milk production [73]. This result was unexpected because epithelial ER is required for lobulo-alveologenesis [67,74,75]. One explanation is the possibility that FOXA1 actively represses maturation of the alveolar lineage, and its loss in virgin epithelia leads to premature alveologenesis independently of ER. To test this possibility, a novel mouse model must be developed wherein FOXA1 expression is enforced during pregnancy. If alveologenesis fails to occur in the presence of pregnancy-associated hormones, this would suggest that FOXA1 plays two, somewhat opposing, roles in the mammary gland. Specifically, it may promote morphogenesis of ducts, but inhibit terminal differentiation of alveoli. Lastly, it is interesting to note that like in the prostate, FOXA2 is not expressed in the mature mammary gland [18]. Thus the mammary gland represents a second tissue where FOXA1 acts independently of its related FOXA member to control morphogenesis.

FOXA1 IN CANCER

Over the past decade, FOXA1 expression has been examined in several human cancers, with proposed oncogenic and tumour suppressive roles depending on the type, and, in some cases, cancer subtype (Table 2). Our knowledge of FOXA1’s participation in four of the cancer types [AML (acute myeloid leukaemia), oesophageal, lung, pancreatic and thyroid] is very much in its infancy with only one or two reports evaluating a role for FOXA1 in each of these diseases. Briefly, FOXA1 is increased in AML patient samples that are chromosomally normal, but harbour oncogenic mutations in FLT3 (fms-related tyrosine kinase 3) and NRAS (neuroblastoma rat sarcoma viral oncogene homologue) [76]. Similarly, the FOXA1 gene is amplified in anaplastic thyroid carcinomas [77], and in a subset of oesophageal and lung cancers [78]. These results suggest that aberrant FOXA1 transcriptional activity may promote tumorigenesis or the level of tumour aggressiveness in these tissues. Further supporting this possibility, FOXA1 expression is correlated with lymph node metastases in ESCC (oesophageal squamous cell carcinoma) and loss of FOXA1 in vitro decreases cellular invasion and migration in an oesophageal cancer cell line [79]. In stark contrast, loss of either FOXA1 or FOXA2 in pancreatic cancer cells induces EMT (epithelial-to-mesenchymal transition), implying a metastasis suppressive role in this disease [80]. The combined knockdown of FOXA1/2 in pancreatic cancer cells leads to an even greater reduction in epithelial-specific gene expression and increased cell motility than silencing either factor alone, suggesting that these genes may co-operate to inhibit metastatic progression. Given the compensatory effect of FOXA2 observed in the pancreas-specific knockout of Foxal during development, it is not surprising that the two FOXA factors have similar functions in cancer cells derived from this tissue. Determining whether loss of either FOXA1 or FOXA2 actually increases metastasis formation in pancreatic cancer mouse models may reveal regulatory mechanisms that modulate this highly aggressive cancer, and lead to the identification of FOXA target genes that could serve as novel therapeutic targets.

Table 2.

Overview of FOXA1 in human cancer

Cancer type FOXA1 expression relative to normal Predicted activity Reference(s)
AML Increased Oncogenic [76]
Breast No difference Unclear [64,65,128132]
Amplified in ER+/PR+ tumours Oncogenic [133]
Oesophageal Amplified/overexpressed Oncogenic [78,79]
HCC Expressed in favourable prognosis tumours Tumour suppressive [85]
Lung Amplified/overexpressed Oncogenic [78]
Pancreatic Decreased in poorly differentiated disease Tumour suppressive [80]
Prostate No difference Unclear [59]
Decreased in associated metastases/favourable outcome Tumour suppressive [110]
Increased in associated metastases/poor outcome Oncogenic [102,103]
Amplified in associated metastases Oncogenic [104]
Thyroid Amplified/overexpressed Oncogenic [77]

Studies investigating the development of the lung, brain and prostate in the absence of FOXA1 have revealed a role for FOXA1 in regulating SHH expression as discussed above. SHH is oncogenic in several cancers [81] including those of the brain, oesophagus, lung and pancreas, all organs for which FOXA1 is either necessary for proper development, or has been implicated in tumorigenesis (Tables 1 and 2). Thus it is feasible that FOXA1 functionally interplays with SHH in one or more of these cancer types to promote initiation and/or progression. Experimental manipulation of FOXA1 expression in established cancer cell lines and transgenic mouse modelling should reveal what role, if any, FOXA1 plays in these cancers. The impact on SHH signalling should also be investigated upon altering FOXA1 expression to determine if FOXA1 mediates tumorigenicity through transcriptional regulation of this pathway. Although FOXA1 has yet to be implicated in CNS tumours, the aberrant up-regulation of Gli1, a transcriptional modulator of SHH, in the developing frog leads to neural tube hyperplasia [82]. Loss of Foxal reduces expression of Gli1/2 in the murine brain [49], thus FOXA1 has the potential to mediate oncogenic SHH signalling in brain cancer. Additionally, although it is clear that FOXA2 compensates for loss of Foxal during development of the brain, lung and pancreas, it will be critical to investigate whether this compensation exists in cancerous cells.

As indicated above, loss of both Foxal and Foxa2 induces bile duct hyperplasia in the murine liver [32]. These results suggest that FOXA proteins may reduce tumour susceptibility, but, interestingly, expression of both FOXA1 and FOXA2 is maintained in chemically induced mouse liver cancer models [83,84]. Although counterintuitive, these results may indicate that FOXA1 is differentially expressed in a less aggressive subtype of HCC (hepatocellular carcinoma) possibly represented in these mouse models. This is supported by results suggesting both FOXA1 and FOXA2 positively regulate mir122, a microRNA expressed in a subset of HCC that is correlated with favourable prognosis [85], and would be similar to the differential expression of FOXA1 seen in the varying subtypes of breast cancer as discussed below. The bile duct hyperplasia observed in the Foxal/2 null liver stems from diminished FOXA1/2 co-operation with GR in the repression of Il6 [32]. Importantly, FOXAs are implicated in GR-mediated transactivation of numerous other target genes [8694], and GR signalling is anti-apoptotic in hepatoma cells [95]. Thus, the potential for pro-tumorigenic FOXA/GR signalling in HCC requires further evaluation. It will be necessary to investigate the expression profile of FOXA1 and FOXA2 in a large cohort of HCC patient samples and compare the presence or absence of FOXA1/2 with aberrant GR/IL-6 signalling to determine whether there is a clinically relevant connection between these pathways.

It is critical to note that while the studies discussed above are suggestive that FOXA1 functions in a pro-tumorigenic or anti-tumorigenic capacity, the rigorous approaches necessary to define FOXA1 as an oncogene (overexpression causes cancer), tumour suppressor (loss causes cancer) or metastasis suppressor (overexpression blocks metastasis) have not been performed for any cancer type. That said, the role for FOXA1 in prostate and breast cancers has been studied to a much greater degree than in any other type of cancer. In these cancers, disease aggressiveness and associated patient prognosis are complicated by multiple cancer subtypes and states of hormone dependence or independence. The known participation of FOXA1 in prostate and breast cancer is detailed below.

FOXA1 in prostate cancer

As previously discussed, FOXA1 is critical for prostate development and loss of Foxa1 causes epithelial hyper-proliferation, activation of oncogenic SHH and decreased expression of the tumour suppressor Nkx3.1 [58]. These results suggest a tumour suppressive role for FOXA1 in prostate epithelia, and pose several questions regarding if, and how, FOXA1 regulates prostatic tumour initiation, progression and/or metastasis. While Foxa1 null prostates maintain epithelial AR expression, they lack AR-dependent secretory differentiation, implying that FOXA1 may modulate AR activation of this process [58]. Given the majority of prostate cancers are driven by aberrant AR signalling, much work has been done to define a role for FOXA1 in this process.

FOXA1 expression in prostate cancer

The LPB-Tag (T antigen) LADY mouse model of prostate cancer expresses SV40 large Tag under control of a large (12 kb) fragment of the prostate-specific probasin promoter (LPB), giving rise to prostate tumours that recapitulate human disease [96]. LADY tumours have increased nuclear expression of FOXA1 and AR that accompanies Tag [59]. Interestingly, FOXA1 is retained in both androgen-dependent (12T-7f) and androgen-independent (12T-10) models. Furthermore, 12T-10-associated liver metastases maintain FOXA1. Together, these results argue against a tumour suppressive role for FOXA1 and suggest that it may function independent of androgens. FOXA1 has also been examined in the TRAMP (transgenic adenocarcinoma of mouse prostate) mouse model that differs from LADY by expressing both SV40 large T- and small t-antigens downstream of a minimal probasin promoter [97,98]. Chiaverotti et al. [99] examined murine strain-specific differences on TRAMP-induced tumour formation and found that expression of the transgene in the FVB/N background decreases survival and increases the incidence of NE carcinomas compared with TRAMP-C57BL/6 animals. NE carcinoma, although rare in humans, is unresponsive to androgen ablation therapy [100]. Interestingly, TRAMP-induced NE carcinomas co-express FOXA1 and FOXA2. Normal prostate epithelia and atypical hyperplasia have high levels of FOXA1 and AR, whereas FOXA2 is only expressed within a small percentage of mature prostate basal epithelial cells [57,59]. This, along with co-expression of epithelial (E-cadherin) and NE (syn-aptophysin) lineage markers, support the possibility that NE carcinomas initiate within a bipotential epithelial population that is more susceptible to tumorigenic processes in FVB/N compared with C57BL/6 mice. The combined expression of FOXA1 and FOXA2 in NE tumours may provide a growth advantage in the absence of androgens. Indeed, in vitro analyses revealed that FOXA2 activates PSA (prostate-specific antigen) promoter activity in an androgen-independent fashion, and suggested that the conversion from FOXA2-negativity into FOXA2-positivity may be responsible for the transition from androgen-dependence to androgen-independence [59].

FOXA1 mRNA positively correlates with human prostate cancer cell line and xenograft models that have been defined as androgen dependent [101]. Examination of FOXA1 and FOXA2 expression in a small cohort of human prostate adenocarcinomas indicated that FOXA1 is present in all samples independent of Gleason score, with the level of expression being indistinguishable from benign tissue [59]. In contrast, FOXA2 is undetectable in low-grade prostate adenocarcinomas, but is found in NE small cell carcinomas similar to the TRAMP mouse model discussed above. The expression of FOXA1 was not reported for these human NE samples [59]. Recent studies investigating FOXA1 protein levels in primary and metastatic human prostate tumours, revealed high FOXA1 expression positively correlates with metastatic disease and poorer outcomes [102,103]. In addition, an amplicon at 14q21, which includes FOXA1, has been identified in metastatic prostate tumours [104]. Lastly, examination of FOXA1 and AR expression revealed poorer outcomes from prostate cancer when these two proteins are co-expressed compared with expression of AR alone [103]. Overall, these studies confirm that FOXA1 expression is maintained in mouse models of prostate cancer as well as human tumours, but more robust expression analyses in human prostate tumours are necessary to delineate its association with patient outcomes and anti-androgen treatment response. In addition, the specific functions that FOXA1 may have in androgen-dependent compared with androgen-independent disease require further exploration. Determining if FOXA1 can be used as a predictor of anti-androgen therapeutic success could provide a valuable diagnostic in the management of prostate cancer.

FOXA1 and the androgen receptor

Most men diagnosed with prostate cancer have AR-positive, hormone-dependent disease that is initially responsive to androgen-ablation therapy. Unfortunately, the majority of these cases become resistant to this therapeutic approach due to acquisition of androgen-independence [105]. To better understand AR signalling within both androgen-dependent and -independent contexts, the co-operation of FOXA1 with AR transactivation has been investigated, albeit with some discrepancies in the resulting data [59,106109]. Initial reports indicated that FOXA1 is necessary for AR activation of target gene expression [106]. These studies involved blockade of FOXA1 binding to the probasin and PSA promoters by mutating the FOXA1 consensus sequences within the promoters. Such disruption blocked androgen (R1881)-induced promoter activity [106], suggesting that FOXA1 is necessary for androgen activation of the expression of these genes (Figure 1). In contrast with these results, overexpression of FOXA1 in other studies inhibited DHT (dihydrotestosterone)-induced PSA promoter activity [59] and AR activation of transcription [109]. While the overexpression of FOXA1 in vitro could non-specifically squelch the transcriptional machinery, and explain these discrepant results, additional studies have shown that reducing FOXA1 expression does not alter DHT-induced expression of PSA/KLK3 (kallikrein-related peptidase 3) [107,108], TMPRSS2 (transmembrane protease serine 2) or PDE9A (phosphodiesterase 9A) [107], nor is FOXA1 necessary for androgen-induced cell cycle progression [107]. However, loss of FOXA1 increases expression of a subset of DHT-induced genes, including PSA/KLK3, and intensifies androgen-induced cell cycle progression [110]. Interestingly, Jia et al. [108] observed a reduction in DHT-induced expression of TMPRSS2 following FOXA1 silencing in an LNCaP variant (C4-2B) that is AR-positive, but androgen-independent, suggesting that FOXA1 facilitates AR-stimulated transcription of at least some genes. Supporting this finding, FOXA1 is also necessary for expression of UBE2C (ubiquitin-conjugating enzyme E2C) in an AR-positive, androgen-independent LNCaP variant (abl), but not in parental cells [111]. FOXA1 also binds to, and may regulate the homeobox transcription factor Hoxbl3 [112], which is a uniquely androgen-independent gene [113]. Overall, these results suggest that FOXA1 modulates AR target gene transcriptional activity, but through various mechanisms that depend on the level of responsiveness of the cell or gene to androgens. They also indicate that FOXA1 can modulate ligand-dependent and -independent actions of AR in prostate cancer cells.

Figure 1. FOXA1/AR signalling in prostate cancer.

Figure 1

Lineage-specific FOXA1-binding sites are marked by histone H3, lysine 4 dimethylation (H3K4me2). Under both androgen-dependent (shown) and androgen-independent (not shown) conditions, FOXA1 mediates AR transactivation of varied responsive genes. FOXA1 and AR also directly interact. Loss of this interaction leads to AR binding and transcriptional regulation at a different set of genes (i.e. hormonal reprogramming) compared with when co-expressed with FOXA1.

Analyses of the binding of FOXA1 to AR target genes and its role in AR transactivation have suggested varied functions for FOXA1 that are gene-specific. FOXA1 binds adjacent to ARE (androgen response elements) in the probasin, PSA [106,114] and Sbp [114] promoters similarly in the presence and absence of DHT. This is similarly seen on a genome-wide level [110]. Others have also reported that FOXA1 binds to AR bound regions, but recruitment of FOXA1 to these sites is enhanced by DHT [107,108]. Importantly, while FOXA1 was necessary for androgen-induced transcriptional activity of the probasin and PSA promoters, blocking direct binding of FOXA1 to these promoters did not impact AR binding [106], suggesting that FOXA1 is not necessary for recruitment of AR. In contrast, AR binding to enhancer regions of several other genes [UBE2C, CDKl (cyclin-dependent kinase 1) and CDC20 (cell division cycle 20)] was reduced in response to FOXA1 silencing in parental (LNCaP) and androgen-independent (LNCaP-abl) prostate cancer cell lines [111]. Hence, the dependency of AR binding on preoccupancy of FOXA1 at adjacent sites appears to be highly dependent on gene context [111]. Supporting the requirement for FOXA1 binding as an essential modulator of AR activity, a germ line SNP (single nucleotide polymorphism) identified in prostate cancer patients within the risk locus 8q24 increases its affinity for FOXA1 binding and potentiates the ability of this chromosomal region to mediate androgen responsiveness of a heterologous promoter compared with the wild-type sequence [115]. Although these results suggest a tight link between FOXA1 binding and androgen responsiveness, the authors did not directly investigate whether FOXA1 is necessary for either AR binding or enhancer activity in its native chromosomal context. As a mechanism for how FOXA1 dictates AR binding at specific genes, studies in both prostate and breast cancer cells revealed that H3K4 dimethylation occurs proximal to FOXA1-binding sites, and can precede FOXA1 binding to DNA [116]. The overexpression of the histone demethylase KDM1 (lysine-specific demethylase 1) reduced AR binding, probably due to loss of bound FOXA1 [111]. The presence of FOXA1 in LNCaP cells is also associated with DNA hypomethylation [13], and an open chromatin state as measured by FAIRE (formaldehyde-assisted isolation of regulatory elements), where high FAIRE FOXA1-binding sites are more likely to bind AR [117]. Overall, it is quite evident that FOXA1 participates in AR target-gene recognition, but it seems likely that this regulation is dependent on additional co-factors, androgen dependence and/or the distance from the transcriptional start site (i.e. promoter or enhancer).

As another mode of regulation, the winged helix/forkhead domain of FOXA1 has been found to complex directly with the DNA-binding domain/hinge region of AR [106,110,118]. This complex permits AR binding to promoter DNA even in the absence of AR consensus elements and vice versa for FOXA1 binding [118]. Formation of a FOXA1/AR multimeric transcriptional complex that requires only one of the two protein’s consensus elements could explain why mutagenesis of FOXA1-binding sites does not alter AR binding [106], whereas decreasing FOXA1 expression does [111] as discussed above. The FOXA1/AR complex may also be necessary for AR nuclear localization. In this case, decreasing FOXA1 expression would greatly impede the access of AR to its target genes, whereas mutating the FOXA1 consensus element would only directly affect FOXA1 binding. Most recently, it has been demonstrated by two distinct groups that the FOXA1/AR interaction is required for maintaining a particular hormonal transcriptional programme [103,110]. Loss of this interaction, either through experimental knockdown of FOXA1, or via somatic mutation of the AR gene, results in an AR-induced transcriptional programme that is distinct from that induced by AR when co-expressed with FOXA1. This is due to the differential binding of AR to targets that are occupied by FOXA1 compared with those that lack a FOXA1-binding site. FOXA1 can also form complexes with USF2 (upstream stimulatory factor 2) in prostate cancer cells, but USF2 does not bind to AR, nor is it required for androgen-induced PSA/KLK3 expression [114]. Since USF2 binds to a similar DNA consensus element as FOXA1, it is also possible that a complex of FOXA1 and USF2 may modulate prostate-specific promoter activity, but this remains to be directly examined.

Additional roles of FOXA1 in prostate cancer

Although the majority of work investigating FOXA1 in prostate cancer has been aimed at understanding how it is involved in modulating AR transcriptional activity, FOXA1 has also been implicated in other signalling pathways. FOXA1/2 positively regulate transcription of the gene AGR2 (anterior gradient protein 2), whose expression increases aggressiveness of prostate cancer cells. This regulation can be abrogated by the presence of EBP1 (ERBB3-binding protein 1) [119]. AGR2 is an established metastasis inducer [120], and its activation by FOXA1 was first discovered in goblet cells [121]. Placement of FOXA1 in the EBP1/FOXA1/AGR2 pathway indicates that it may play an important modulatory role in metastatic progression. Interestingly, EBP1 has also been shown to negatively regulate AR-mediated transcription [122]. While not specifically tested, it is possible that EBP1 represses AR transactivation by inhibiting FOXA1 function in a similar manner as seen for the transcriptional regulation of AGR2. In contrast to its pro-metastatic positive regulation of AGR2, expression of FOXA1 is also down-regulated by SOX4 (sex determining region Y-box 4), a prostate cancer oncogene [123]. This contradiction exemplifies the broad spectrum of activity for FOXA1 in the cancerous prostate, and at least partially explains how loss of Foxa1 in the normal prostate could lead to both tumorigenic (i.e. increased SHH) and anti-tumorigenic (i.e. loss of AR function) phenotypes. Lastly, it is intriguing to speculate a potential pro-carcinogenic functional interaction between FOXA1, Nkx3.1 and SHH in the prostate given the aberrant regulation of these three factors in the Foxa1 null prostate. Future in vitro work should aim to delineate the connection between these pathways and their interdependent contributions to disease phenotypes.

FOXA1 in breast cancer

The molecular subtypes of breast cancer were identified via unsupervised hierarchical clustering of cDNA microarray data from a cohort of human breast tumours, in which each subtype was distinguished by the differential expression of numerous genes. In particular, FOXA1 mRNA is expressed in luminal subtype tumours along with several other discriminatory genes, including ESR1 and GATA3 [124,125]. While the six distinct subtypes [luminal A, luminal B, luminal C, HER2/ERBB2, basal and normal-like] are classified by genome-wide expression profiling [125], they are often identified using the surrogate markers: ER, PR (progesterone receptor) and the epidermal growth factor receptor 2 (HER2/ERBB2), a proto-oncogene amplified in ~ 20–30% of breast cancer patients. Luminal A subtype tumours are ER+ /PR+ /HER2 and confer a favourable prognostic outcome that is at least partially due to the efficacy of anti-hormone therapies [126]. In contrast, basal subtype tumours tend to be triple negative (ER /PR /HER2) and confer a poor prognosis due to the inherent aggressiveness of these tumours and the absence of subtype-specific targeted therapies [127].

FOXA1 expression in breast cancer

Since being identified as a novel luminal subtype gene, the clinical significance of FOXA1 protein expression in breast tumours has been investigated by multiple groups. Wolf et al. [128] were the first to investigate a breast tumour TMA (tissue microarray) for FOXA1 expression, revealing that FOXA1 is associated with low tumour grade, and, not surprisingly, ER expression [129]. A number of publications soon followed with various TMA compositions, and, when combined, provide expression data for FOXA1 on over 5000 human breast cancers [64,65,129132]. FOXA1 significantly associates with ER expression in each study. Moreover, FOXA1 positively correlates with the luminal subtype as defined by ER and/or PR positivity, HER2 negativity [64,129], and luminal CK expression [64,65,129]. Negative correlations have been observed for FOXA1 and the basal subtype when defined as ER-negative, HER2-negative with CK5/6 and/or EGFR positivity [129, 132] or by basal CK expression [65,129]. Importantly, several groups have proposed utilizing FOXA1 as a prognostic tool to stratify patients within the ER-positive luminal subtype [64,129,130]. Results from these studies suggest that FOXA1 can predict survival within this population, and thus aid in therapeutic decision-making. However, TMA data generated by one group argues that FOXA1 is unable to further stratify prognosis within the subgroup of patients with ER-positive disease [65]. To directly test the independent prognostic and predictive ability of FOXA1, Ademuyiwa et al. [131] investigated the correlation between FOXA1 expression and Oncotype DX recurrence scores in patients with ER-positive node-negative disease. The Oncotype DX is a molecular diagnostic used to predict recurrence and response to chemotherapy. FOXA1 negatively correlated with recurrence score leading the authors to suggest FOXA1 immunostaining could function as a more cost-effective pathological marker than the Oncotype DX. Of note, FOXA1 is not a component of Oncotype DX. Lastly, increased copy number of chromosomal locus 14q13, where FOXA1 is located, is associated with the ER- and PR-positive breast cancer subtype [133], providing further evidence that FOXA1 may promote tumour differentiation.

Although FOXA1 statistically correlates with ER, it has been noted that some ER-negative tumours also have high FOXA1 expression [65,128]. In fact, a subset of ER- and PR-negative tumours is molecularly more similar to ER- and PR-positive than to triple negative tumours. Most importantly, FOXA1 was identified as one of the differentially expressed genes within this ‘ER-positive-like’ subgroup [134]. These tumours are more likely to possess apocrine features, express AR and have an androgen responsive molecular signature, and a breast cancer cell line (MDA-MB-453) that recapitulates this phenotype is growth stimulated by androgens and inhibited by anti-androgens [135]. Given the role of FOXA1 in modulating AR transactivation in prostate cancer, it is possible that the presence of FOXA1 in these ER-negative tumours is required for androgen-dependent responsiveness mediated by AR. Supporting this hypothesis, testosterone increases FOXA1 mRNA expression in another AR-positive breast cancer cell line (SUM190) [136]. Indeed, recent studies provide evidence that FOXA1 mediates AR binding and transcriptional regulation in the MDA-MB-453 cells [137,138]. FOXA1/AR signalling in these cells is similar to FOXA1/ER signalling in MCF7 cells, and is necessary for maintenance of the apocrine gene expression signature [137]. Further evidence revealed that AR transcriptionally regulates WNT7B, and subsequent induction of HER3 gene expression via an AR/FOXA1/β-catenin complex [138]. Enhanced HER3/HER2 signalling likely drives tumorigenesis of apocrine tumors. Hence, FOXA1/AR induces a set of genes that convey the phenotype, and perhaps induces the formation, of apocrine of breast cancers.

FOXA1 and the oestrogen receptor

Functioning as a chromatin remodeller, FOXA1 was discovered to co-operate with ER in activating the liver-specific vitellogenin promoter [139], and had been shown to regulate the well-established ER target, TFF1 (trefoil factor 1; pS2) [140]. In 2005, two groups independently extended these findings to breast cancer, demonstrating that FOXA1 was necessary for oestrogen-induced ER binding, and the up-regulation of several target genes, including TFF1 [141,142]. Carrolletal. [141] identified forkhead consensus motifs in close proximity to approximately half of the ER-binding sites spanning chromosomes 21 and 22 of a breast cancer cell line. Likewise, Laganière et al. [142] analysed ER-binding sites via a human promoter array, and found 12% of ER-bound promoters also contained forkhead motifs [143]. Several genome-wide analyses have confirmed these data [116,143146], although one group has reported much less (<10%) commonality in the location of forkhead and ER-binding motifs in the genome [147]. That said, considerable co-operativity between FOXA1 and ER has been repeatedly observed in comparisons of full genome ER and FOXA1 ChlP-chip [116] and ChlP-seq [146] data, showing that over half of ER-binding sites are also occupied by FOXA1. This co-operation is observed for both ER up-regulated and down-regulated genes [116,146] (Figure 2A). For example, FOXA1 is necessary for ER repression of Reprimo (RPRM) transcription [148]. FOXA1 and ER can also have opposing roles in regulating gene expression as demonstrated for BASE (breast cancer and salivary gland expression); FOXA1 is required for BASE expression, whereas ER represses transcription of this gene [149].

Figure 2. FOXA1/ER signalling in breast cancer.

Figure 2

(A) Lineage-specific FOXA1-binding sites are marked by histone H3, lysine 4 dimethylation (H3K4me2). FOXA1 mediates both oestrogen-induced gene transactivation and repression. (B) FOXA1 is necessary for ER expression, and expression of ER has been proposed to modulate FOXA1 expression in an oestrogen-dependent manner.

Functionally, FOXA1 is necessary for cell growth [150] and specifically for oestrogen-mediated cell-cycle progression of breast cancer cells [142,151]. These results are at least partly explained by the requirement for FOXA1 to mediate ER stimulation of CCND1, the gene encoding cyclin D1, through a downstream enhancer [151]. The presence of FOXA1 correlates with bound RNA polymerase II, active chromatin and histone H4 acetylation [151] at ER-activated genes, where each is a hallmark of proliferating cells. Similar to its role in prostate cancer cells, FOXA1 binding is associated with an open chromatin state as measured by FAIRE, where loss of FOXA1 decreases the level of open chromatin on a global scale [117,146]. Specifically, FOXA1 binding at high FAIRE regions is accompanied by increased histone H3, lysine 9 (H3K9) acetylation, decreased H3K9 mono- and dimethylation, and increased H3K4 dimethylation [117]. The presence of FOXA1 is also associated with DNA hypomethylation [13]. Together, these results support a pioneering function for FOXA1 in modulating chromatin structure for subsequent ER binding and activity (Figure 2A), similar to that predicted for AR, although the specific timing of events is not well established in cancer cell lines. In addition to modulating ER activity, we recently reported that FOXA1 also directly binds to the ESR1 (oestrogen receptor 1) promoter and is required for expression of ER mRNA and protein in breast cancer cells [73]. Thus, unlike its relationship with AR in the prostate, FOXA1 controls both the expression and activity of ER in breast cancer (Figure 2B).

In response to oestrogen stimulation, FOXA1 is preferentially bound to intergenic enhancers and introns, mirroring that of ER [141]. It is recruited to DNA in the presence and absence of estradiol, but unlike with androgen stimulation, binding of FOXA1 generally decreases with oestrogen treatment [141,143]. A direct comparison of FOXA1 in ER and AR signalling using breast and prostate cells, respectively, revealed that while there are common binding sites, FOXA1 has a distinct binding profile depending on whether it is co-expressed with ER or AR [116]. This tissue specificity is dependent on differential recruitment of FOXA1 to ER compared with AR target genes due to lineage-specific H3K4 dimethylation [116]. Moreover, the presence of FOXA1 at the TFF1 gene enhancer is also dependent on binding of the histone variant H2A.Z by p400 [152], providing further evidence that tissue-specific FOXA1 activity is dictated by histone modifications. Another factor has recently been identified that also delineates FOXA1-specific recruitment and subsequent ER transcriptional regulation. CTCF (CCCTC-binding factor) is an insulator binding protein [153,154] whose loss was shown to impede FOXA1 binding to known target genes [e.g. TFF1 and PGR (progesterone receptor)], while loss of FOXA1 had no impact on CTCF binding [154]. In contrast, Hurtado et al. [146] found that loss of CTCF increases FOXA1-binding capacity. Despite the discrepancy between these two reports, it is apparent that CTCF can modulate FOXA1 binding. The context dependency of this interaction must be further explored to unravel the molecular mechanisms by which CTCF enhances or impedes binding of FOXA1 to DNA.

The interplay between FOXA1 and ER may extend beyond the ability of FOXA1 to control ER expression and activity. Using a different experimental approach than Carroll et al. [141] who identified FOXA1-binding sites in ER target genes through motif analysis, Laganière et al. [142] first identified FOXA1 as an oestrogen-induced ER target gene and then subsequently observed FOXA1-binding sites within ER responsive genes. Providing support for FOXA1 being an ER target, a more recent analysis of the ER chromatin interaction network revealed that ER-binding sites flank the FOXA1 gene [155]. Although these results suggest a positive regulatory loop between ER and FOXA1 (Figure 2B), others have described FOXA1 to be down-regulated with oestrogen treatment [128,156158]. Moreover, mice-lacking Esr1 maintain expression of FOXA1 in the mammary epithelium [73]. These disparate observations are further complicated by the varying roles of FOXA1 in mediating ER signalling in tissues other than the breast [146,159,160]. Osteosarcoma cells that have been engineered to express the ER (U2OS-ER) have undetectable levels of FOXA1 protein, suggesting that the ER is insufficient to induce expression of FOXA1. Given the absence of FOXA1 in U2OS-ER cells, it is not surprising that forkhead consensus motifs do not correlate with the ER cistrome, nor is FOXA1 required for ER-regulated transcription in this context [160]. These results suggest that the role of FOXA1 in modulating ER activity is context dependent, and are consistent with the observation that FOXA1 is necessary for ER expression only in the epithelium and not in the stroma of the murine mammary gland [73]. In support of an indispensable role for FOXA1 in ER activity in breast epithelial-specific gene regulation, exogenous expression of FOXA1 in U2OS-ER cells induces ER binding to otherwise breast-specific ER targets [e.g. TFF1 and XBP1 (X-box binding protein 1)] that is accompanied by increased expression of these genes [146]. Furthermore, FOXA1 overexpressing U2OS-ER cells also become sensitive to the growth inhibitory effects of tamoxifen, a SERM (selective ER modulator). In contrast, FOXA1 is endogenously expressed in, and is necessary for, resveratrol-induced ER transactivation of BMP2 (bone morphogenetic protein-2) gene expression in the osteoblast cell line MC3T3-E1 [159]. It is possible that the distinct observations seen with FOXA1 and ER in these studies can be attributed to the differences between the U2OS cells that were derived from a human osteosarcoma compared with the non-transformed MC3T3-E1 cells that were isolated from newborn mouse calvaria [161]. FOXA1 has been proposed as a putative breast cancer therapeutic target based on the concept that ER dependency on FOXA1 will translate to an increase in the efficacy of tamoxifen in ER-dependent breast cancer [162,163]. Functioning as an SERM, tamoxifen has antagonistic properties in the breast, but is agonistic in the bone, where it maintains oestrogen signalling and is protective against osteoporosis. The differing roles of FOXA1 in cancerous and normal bone tissue should be further defined before considering targeting FOXA1 in breast cancer patients. If FOXA1 is expressed in normal bone, as seen in the MC3T3-E1 cells, but loss of FOXA1 correlates with tumorigenic progression, as shown by the lack of FOXA1 in transformed U2OS cells, then the systemic reduction of FOXA1 expression could inadvertently induce bone tumours.

Additional roles of FOXA1 in breast cancer

In addition to the well-described ability of FOXA1 to control ER activity, there have been other facets of FOXA1 described in breast cancer. One of these is a possible role in HER2 signalling [136,150]. Treatment of the breast cancer cell line SUM190 with heregulin, an indirect activator of HER2, induces FOXA1 expression [136]. This suggests that FOXA1 may be downstream of HER2 signalling and mediate some of its actions. Indeed, FOXA1 silencing potentiates the cellular toxicity of herceptin, a humanized monoclonal antibody that inactivates HER2 in HER2-amplified breast cancer cell lines [150]. While the specific mechanisms remain unknown, these results support a role for FOXA1 in HER2-induced cell survival. However, it is important to note that the presence or absence of FOXA1 does not correlate with clinical outcome of patients with HER2-positive disease [164], indicating that FOXA1 may not play a critical role in HER2-induced signalling in vivo.

In addition to many other genes, FOXA1 binds to, and activates transcription of, the gene encoding HSP72 (heat-shock protein 72; HSPA1A), the stress-inducible cytosolic form of HSP70 [165]. HSP70 independently associates with poorer outcome in breast cancer patients with node-negative disease [166]. Thus it is possible that FOXA1 and HSP72 co-operatively dictate the aggressive behaviours of these cancers, but this has not been tested. As mentioned earlier, FOXA1 also participates in ER-dependent transactivation of CCND1 (cyclin D1) [151]. Intriguingly, FOXA1, ER and cyclin D1 are co-ordinately up-regulated in tumours arising in mice that are bi-transgenic for mammary gland-specific expression of ILK (integrin-linked kinase) and Wnt1 (MMTV-Wnt1/ILK) [167]. Forced co-expression of ILK and Wnt1 leads to increased epithelial proliferation, a greater percentage of luminal progenitor cells and increased tumour formation as opposed to the expression of either alone. The authors of this work proposed that the convergence of Wnt1 and ILK on β-catenin activation may result in up-regulation of FOXA1, which is downstream of SOX17/β-catenin during endodermal development in Xenopus [168]. Notably we have shown that FOXA1 is expressed in the mammary epithelial luminal progenitor population [73]. Whether FOXA1 is necessary for expansion of luminal progenitors and the formation of mammary tumours, or is simply expressed as a bystander in this bi-transgenic or other mouse models of breast cancer, remains to be seen.

Given its correlation with the less aggressive luminal subtype of breast cancer, it is not surprising that FOXA1 activates the transcription of genes [e.g. CDH1 (t-cadherin) and CDKN1B (cyclin-dependent kinase inhibitor 1B)] implicated in decreased breast cancer tumorigenicity independently of ER [169,170]. Exogenous overexpression of FOXA1 in a triple negative, basal breast cancer cell line (MDA-MB-231) induces expression of the cell-cell adhesion molecule, E-cadherin [171]. In this context, FOXA1 directly stimulates transcription of the E-cadherin gene (CDH1), and the associated induction of E-cadherin expression decreases the migratory capacity of these cells, suggesting that FOXA1 may promote their differentiation. Activation of CDH1 occurs in the absence of ER, supporting the notion that FOXA1 has ER-independent roles in dictating a more differentiated luminal cell phenotype. Another example of FOXA1 functioning in a tumour suppressive role is its regulation of the cyclin-dependent kinase inhibitor p27Kip1 [172]. FOXA1 binds to the promoter of p27Kip1 (CDKNlB) in breast cancer cells and synergizes with BRCA1 (breast-cancer susceptibility gene 1) to induce its transactivation in a colon cancer cell line. In addition, breast cancer cells overexpressing FOXA1 have increased p27Kip1 promoter activity and decreased cell number [128]. When combined, these studies paint a complicated picture of FOXA1 as a participant in multiple signalling pathways in breast cancer, which are both oncogenic and tumour suppressive. It is likely that the specific molecular functions of FOXA1 are dictated by many components, including, but not limited to, the epithelial origin of the tumour initiating cell and the co-expression of ER, HER2 and other regulatory factors that have not yet been identified.

CONCLUDING REMARKS

Since first described in liver-specific transcription in 1989, the FOXA subfamily has been linked to the development and differentiation of multiple tissues. FOXA1, in particular, has roles in the pancreas, kidney, liver, lung, gastro-intestinal tract, brain, prostate and mammary gland, and will likely prove to be expressed in, and modulate the function of, additional tissues. Commonly, FOXA1 is necessary for epithelial secretion in several organs, and is analogously critical in mediating lineage specification. FOXA2 can compensate for loss of Foxa1 in the development of many tissues, but in others (e.g. mammary, prostate) the lack of redundancy can be, at least partly, explained by the lineage-specific expression of FOXA1 in the absence of FOXA2. Similarly, expression of FOXA3 is undetectable in the murine prostate [57] and in breast cancer cell lines [13]. Beyond having divergent expression profiles, the apparent lack of compensation by FOXA3 for either FOXA1 or FOXA2 is likely due to the dissimilarity of the amino acid sequence of this factor with FOXA1 and FOXA2 outside of the forkhead domain [4], and their distinct DNA-binding profiles [4,173]. To truly test whether FOXA3 is capable of compensating for loss of Foxa1/2, mice null for both Foxa1/a2 and Foxa3 will be necessary.

Developmental studies have revealed FOXA1 regulates certain genes indiscriminate of cell type (e.g. Shh), while other FOXA1 targets are tissue-specific. The pattern of SHH regulation by FOXA1 is also tissue specific, where loss of Foxa1 decreases SHH in the lung and brain, but increases SHH in the prostate. The interplay between FOXA1 and SHH requires further evaluation given the presence of aberrant SHH signalling in multiple types of cancer. In addition, FOXA1 has a propensity to co-operate with nuclear hormone receptors (e.g. GR, ER and AR) in a somewhat similar manner in both development and cancer. The ability of FOXA1 to modulate AR and ER transcriptional regulation has been studied in the greatest detail in prostate and breast cancer respectively, but has been primarily limited to hormone-dependent disease. It will be critical to further investigate the potential role of FOXA1 in the transition to hormone independence and in hormone receptor negative disease, both features of more aggressive, less treatable cancers. The ‘cell of origin’ hypothesis predicts that a tumour maintains the molecular characteristics of the cell where the cancer originated. With regard to the breast, multiple subtypes of cancer exist representing the distinct lineages of the normal mammary epithelium, and these subtypes are predictive of patient prognosis. In this disease FOXA1 is expressed only in the luminal subtype, and is predictive of favourable patient prognosis. The differential expression and requirement for FOXA1 in the discrete lineages of tissue types other than breast will probably prove an equally valuable subtype-specific marker in cancers associated with these tissues, and may also be predictive of patient prognosis. In particular, the Foxa1 null prostate exhibits a decrease in the luminal lineage, with a concomitant increase in the basal lineage.

In summary, the investigation of Foxa1 knockout mice coupled with cell line studies has uncovered FOXA1-mediated pathways that, in addition to being necessary for normal development, are aberrant in cancer. The majority of our knowledge of FOXA1 in cancer relates to its function in hormone receptor driven breast and prostate cancers. While FOXA1 in hormone signalling should continue to be investigated, future research should also revolve around elucidating a role for FOXA1 in hormone independent breast and prostate cancers, and cancers of other tissues where FOXA1 has been implicated in normal development.

FUNDING

Our own work on FOXA1 was supported by the Department of Defense [grant numbers W81XWH-08-1-0347 (to R.A.K.) and W81XWH-06-1-0712 (to G.M.B.)].

Abbreviations used:

AGR2

anterior gradient protein 2

AML

acute myeloid leukaemia

AR

androgen receptor

ARE

androgen response elements

α-SMA

α-smooth muscle actin

BASE

breast cancer and salivary gland expression

CCSP

clara cell secretory protein

Chip

chromatin immunoprecipitation

CK

cytokeratin

CNS

central nervous system

CTCF

CCCTC-binding factor

DHT

dihydrotestosterone

E

embryonic day

EBP1

ERBB3-binding protein 1

EN1

engrailed 1

ER

oestrogen receptor

FAIRE

formaldehyde-assisted isolation of regulatory elements

FOG2

friend of GATA2

FOX

forkhead box

GR

glucocorticoid receptor

HCC

hepatocellular carcinoma

HNF

hepatocyte nuclear factor

HSP

heat shock protein

IL-6

interleukin-6

ILK

integrin-linked kinase

KLK3

kallikrein-related peptidase 3

MMTV

murine-mammary-tumour virus

NE

neuroendocrine

PDX-1

pancreas and duodenal homeobox protein-1

P

postnatal day

PR

progesterone receptor

PSA

prostate-specific antigen

Sbp

spermine-binding protein

SERM

selective ER modulator

SHH

sonic hedgehog

SP-B

pro-surfactant protein-B

SV40

simian virus 40

Tag

T antigen

TFF1

trefoil factor 1

TMA

tissue microarray

TMPRSS2

transmembrane protease serine 2

TRAMP

transgenic adenocarcinoma of mouse prostate

UBE2C

ubiquitin-conjugating enzyme E2C

Ucp2

uncoupling protein 2

USF2

upstream stimulatory factor 2

REFERENCES

  • 1.Hannenhalli S and Kaestner KH (2009) The evolution of Fox genes and their role in development and disease. Nat. Rev. Genet 10, 233–240 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Costa RH, Grayson DR and Darnell JE Jr (1989) Multiple hepatocyte-enriched nuclear factors function in the regulation of transthyretin and α1-antitrypsin genes. Mol. Cell. Biol 9, 1415–1425 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Kaestner KH (2010) The FoxA factors in organogenesis and differentiation. Curr. Opin. Genet. Dev 20, 527–532 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Lai E, Prezioso VR, Tao WF, Chen WS and Darnell JE Jr (1991) Hepatocyte nuclear factor 3α belongs to a gene family in mammals that is homologous to the Drosophila homeotic gene fork head. Genes Dev. 5, 416–427 [DOI] [PubMed] [Google Scholar]
  • 5.Weigel D, Jurgens G, Kuttner F, Seifert E and Jackle H (1989) The homeotic gene fork head encodes a nuclear protein and is expressed in the terminal regions of the Drosophila embryo. Cell 57, 645–658 [DOI] [PubMed] [Google Scholar]
  • 6.Pani L, Overdier DG, Porcella A, Qian X, Lai E and Costa RH (1992) Hepatocyte nuclear factor 3β contains two transcriptional activation domains, one of which is novel and conserved with the Drosophila fork head protein. Mol. Cell. Biol 12, 3723–3732 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Qian X and Costa RH (1995) Analysis of hepatocyte nuclear factor-3β protein domains required for transcriptional activation and nuclear targeting. Nucleic Acids Res. 23, 1184–1191 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Clark KL, Halay ED, Lai E and Burley SK (1993) Co-crystal structure of the HNF-3/fork head DNA-recognition motif resembles histone H5. Nature 364, 412–420 [DOI] [PubMed] [Google Scholar]
  • 9.Overdier DG, Porcella A and Costa RH (1994) The DNA-binding specificity of the hepatocyte nuclear factor 3/forkhead domain is influenced by amino-acid residues adjacent to the recognition helix. Mol. Cell. Biol 14, 2755–2766 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Cirillo LA, McPherson CE, Bossard P, Stevens K, Cherian S, Shim EY, Clark KL, Burley SK and Zaret KS (1998) Binding of the winged-helix transcription factor HNF3 to a linker histone site on the nucleosome. EMBO J. 17, 244–254 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Cirillo LA and Zaret KS (1999) An early developmental transcription factor complex that is more stable on nucleosome core particles than on free DNA. Mol. Cell 4, 961–969 [DOI] [PubMed] [Google Scholar]
  • 12.Cirillo LA, Lin FR, Cuesta I, Friedman D, Jarnik M and Zaret KS (2002) Opening of compacted chromatin by early developmental transcription factors HNF3 (FoxA) and GATA-4. Mol. Cell 9, 279–289 [DOI] [PubMed] [Google Scholar]
  • 13.Serandour AA, Avner S, Percevault F, Demay F, Bizot M, Lucchetti-Miganeh C, Barloy-Hubler F, Brown M, Lupien M, Metivier R et al. (2011) Epigenetic switch involved in activation of pioneer factor FOXA1-dependent enhancers. Genome Res. 21, 555–565 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Ang SL, Wierda A, Wong D, Stevens KA, Cascio S, Rossant J and Zaret KS (1993) The formation and maintenance of the definitive endoderm lineage in the mouse: involvement of HNF3/forkhead proteins. Development 119, 1301–1315 [DOI] [PubMed] [Google Scholar]
  • 15.Monaghan AP, Kaestner KH, Grau E and Schutz G (1993) Postimplantation expression patterns indicate a role for the mouse forkhead/HNF-3α, β and γ genes in determination of the definitive endoderm, chordamesoderm and neuroectoderm. Development 119, 567–578 [DOI] [PubMed] [Google Scholar]
  • 16.Sasaki H and Hogan BL (1993) Differential expression of multiple fork head related genes during gastrulation and axial pattern formation in the mouse embryo. Development 118, 47–59 [DOI] [PubMed] [Google Scholar]
  • 17.Kaestner KH, Hiemisch H, Luckow B and Schutz G (1994) The HNF-3 gene family of transcription factors in mice: gene structure, cDNA sequence, and mRNA distribution. Genomics 20, 377–385 [DOI] [PubMed] [Google Scholar]
  • 18.Besnard V, Wert SE, Hull WM and Whitsett JA (2004) Immunohistochemical localization of Foxa1 and Foxa2 in mouse embryos and adult tissues. Gene Expr. Patterns 5, 193–208 [DOI] [PubMed] [Google Scholar]
  • 19.Friedman JR and Kaestner KH (2006) The Foxa family of transcription factors in development and metabolism. Cell. Mol. Life Sci. 63, 2317–2328 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Weinstein DC, Ruiz i. Altaba A, Chen WS, Hoodless P, Prezioso VR, Jessell TM and Darnell JE Jr (1994) The winged-helix transcription factor HNF-3β is required for notochord development in the mouse embryo. Cell 78, 575–588 [DOI] [PubMed] [Google Scholar]
  • 21.Kaestner KH, Katz J, Liu Y, Drucker DJ and Schutz G (1999) Inactivation of the winged helix transcription factor HNF3α affects glucose homeostasis and islet glucagon gene expression in vivo. Genes Dev. 13, 495–504 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Shih DQ, Navas MA, Kuwajima S, Duncan SA and Stoffel M (1999) Impaired glucose homeostasis and neonatal mortality in hepatocyte nuclear factor 3α-deficient mice. Proc. Natl. Acad. Sci. U.S.A 96, 10152–10157 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Kaestner KH, Hiemisch H and Schutz G (1998) Targeted disruption of the gene encoding hepatocyte nuclear factor 3γ results in reduced transcription of hepatocyte-specific genes. Mol. Cell. Biol 18, 4245–4251 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Shen W, Scearce LM, Brestelli JE, Sund NJ and Kaestner KH (2001) Foxa3 (hepatocyte nuclear factor 3γ) is required for the regulation of hepatic GLUT2 expression and the maintenance of glucose homeostasis during a prolonged fast. J. Biol. Chem 276, 42812–42817 [DOI] [PubMed] [Google Scholar]
  • 25.Behr R, Sackett SD, Bochkis IM, Le PP and Kaestner KH (2007) Impaired male fertility and atrophy of seminiferous tubules caused by haploinsufficiency for Foxa3. Dev. Biol 306, 636–645 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Vatamaniuk MZ, Gupta RK, Lantz KA, Doliba NM, Matschinsky FM and Kaestner KH (2006) Foxa1-deficient mice exhibit impaired insulin secretion due to uncoupled oxidative phosphorylation. Diabetes 55, 2730–2736 [DOI] [PubMed] [Google Scholar]
  • 27.Gao N, LeLay J, Vatamaniuk MZ, Rieck S, Friedman JR and Kaestner KH (2008) Dynamic regulation of Pdx1 enhancers by Foxa1 and Foxa2 is essential for pancreas development. Genes Dev. 22, 3435–3448 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Gu G, Dubauskaite J and Melton DA (2002) Direct evidence for the pancreatic lineage: NGN3 + cells are islet progenitors and are distinct from duct progenitors. Development 129, 2447–2457 [DOI] [PubMed] [Google Scholar]
  • 29.Gao N, Le Lay J, Qin W, Doliba N, Schug J, Fox AJ, Smirnova O, Matschinsky FM and Kaestner KH (2010) Foxa1 and Foxa2 maintain the metabolic and secretory features of the mature β-cell. Mol. Endocrinol 24, 1594–1604 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Behr R, Brestelli J, Fulmer JT, Miyawaki N, Kleyman TR and Kaestner KH (2004) Mild nephrogenic diabetes insipidus caused by Foxa1 deficiency. J. Biol. Chem 279, 41936–41941 [DOI] [PubMed] [Google Scholar]
  • 31.Lee CS, Friedman JR, Fulmer JT and Kaestner KH (2005) The initiation of liver development is dependent on Foxa transcription factors. Nature 435, 944–947 [DOI] [PubMed] [Google Scholar]
  • 32.Li Z, White P, Tuteja G, Rubins N, Sackett S and Kaestner KH (2009) Foxa1 and Foxa2 regulate bile duct development in mice. J. Clin. Invest 119, 1537–1545 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Bochkis IM, Rubins NE, White P, Furth EE, Friedman JR and Kaestner KH (2008) Hepatocyte-specific ablation of Foxa2 alters bile acid homeostasis and results in endoplasmic reticulum stress. Nat. Med 14, 828–836 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Besnard V, Wert SE, Kaestner KH and Whitsett JA (2005) Stage-specific regulation of respiratory epithelial cell differentiation by Foxa1. Am. J. Physiol. Lung Cell Mol. Physiol 289, L750–L759 [DOI] [PubMed] [Google Scholar]
  • 35.Sawaya PL and Luse DS (1994) Two members of the HNF-3 family have opposite effects on a lung transcriptional element; HNF-3α stimulates and HNF-3β inhibits activity of region I from the Clara cell secretory protein (CCSP) promoter. J. Biol. Chem 269, 22211–22216 [PubMed] [Google Scholar]
  • 36.Clevidence DE, Overdier DG, Peterson RS, Porcella A, Ye H, Paulson KE and Costa RH (1994) Members of the HNF-3/forkhead family of transcription factors exhibit distinct cellular expression patterns in lung and regulate the surfactant protein B promoter. Dev. Biol 166, 195–209 [DOI] [PubMed] [Google Scholar]
  • 37.Bohinski RJ, Di Lauro R and Whitsett JA (1994) The lung-specific surfactant protein B gene promoter is a target for thyroid transcription factor 1 and hepatocyte nuclear factor 3, indicating common factors for organ-specific gene expression along the foregut axis. Mol. Cell. Biol 14, 5671–5681 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Wan H, Dingle S, Xu Y, Besnard V, Kaestner KH, Ang SL, Wert S, Stahlman MT and Whitsett JA (2005) Compensatory roles of Foxa1 and Foxa2 during lung morphogenesis. J. Biol. Chem 280, 13809–13816 [DOI] [PubMed] [Google Scholar]
  • 39.Bellusci S, Furuta Y, Rush MG, Henderson R, Winnier G and Hogan BL (1997) Involvement of Sonic hedgehog (Shh) in mouse embryonic lung growth and morphogenesis. Development 124, 53–63 [DOI] [PubMed] [Google Scholar]
  • 40.Pepicelli CV, Lewis PM and McMahon AP (1998) Sonic hedgehog regulates branching morphogenesis in the mammalian lung. Curr. Biol 8, 1083–1086 [DOI] [PubMed] [Google Scholar]
  • 41.Song L, Zhang B, Feng Y, Luo X, Wei X and Xiao X (2009) A role for forkhead box A1 in acute lung injury. Inflammation 32, 322–332 [DOI] [PubMed] [Google Scholar]
  • 42.Song L, Wei X, Zhang B, Luo X, Liu J, Feng Y and Xiao X (2009) Role of Foxa1 in regulation of bcl2 expression during oxidative-stress-induced apoptosis in A549 type II pneumocytes. Cell Stress Chaperones 14, 417–425 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Ang SL and Rossant J (1994) HNF-3β is essential for node and notochord formation in mouse development. Cell 78, 561–574 [DOI] [PubMed] [Google Scholar]
  • 44.Ferri AL, Lin W, Mavromatakis YE, Wang JC, Sasaki H, Whitsett JA and Ang SL (2007) Foxa1 and Foxa2 regulate multiple phases of midbrain dopaminergic neuron development in a dosage-dependent manner. Development 134, 2761–2769 [DOI] [PubMed] [Google Scholar]
  • 45.Zetterstrom RH, Solomin L, Jansson L, Hoffer BJ, Olson L and Perlmann T (1997) Dopamine neuron agenesis in Nurr1-deficient mice. Science 276, 248–250 [DOI] [PubMed] [Google Scholar]
  • 46.Simon HH, Saueressig H, Wurst W, Goulding MD and O’Leary DD (2001) Fate of midbrain dopaminergic neurons controlled by the engrailed genes. J. Neurosci 21, 3126–3134 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Andersson E, Jensen JB, Parmar M, Guillemot F and Bjorklund A (2006) Development of the mesencephalic dopaminergic neuron system is compromised in the absence of neurogenin 2. Development 133, 507–516 [DOI] [PubMed] [Google Scholar]
  • 48.Lin W, Metzakopian E, Mavromatakis YE, Gao N, Balaskas N, Sasaki H, Briscoe J, Whitsett JA, Goulding M, Kaestner KH et al. (2009) Foxa1 and Foxa2 function both upstream of and cooperatively with Lmx1a and Lmx1b in a feedforward loop promoting mesodiencephalic dopaminergic neuron development. Dev. Biol 333, 386–396 [DOI] [PubMed] [Google Scholar]
  • 49.Mavromatakis YE, Lin W, Metzakopian E, Ferri AL, Yan H, Sasaki H, Whisett J and Ang SL (2011) Foxa1 and Foxa2 positively and negatively regulate Shh signalling to specify ventral midbrain progenitor identity. Mech. Dev 128, 90–103 [DOI] [PubMed] [Google Scholar]
  • 50.Ye DZ and Kaestner KH (2009) Foxa1 and Foxa2 control the differentiation of goblet and enteroendocrine L-and D-cells in mice. Gastroenterology 137, 2052–2062 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.van der Sluis M, Vincent A, Bouma J, Korteland-Van Male A, van Goudoever JB, Renes IB and Van Seuningen I (2008) Forkhead box transcription factors Foxa1 and Foxa2 are important regulators of Muc2 mucin expression in intestinal epithelial cells. Biochem. Biophys. Res. Commun 369, 1108–1113 [DOI] [PubMed] [Google Scholar]
  • 52.Gauthier BR, Schwitzgebel VM, Zaiko M, Mamin A, Ritz-Laser B and Philippe J (2002) Hepatic nuclear factor-3 (HNF-3 or Foxa2) regulates glucagon gene transcription by binding to the G1 and G2 promoter elements. Mol. Endocrinol 16, 170–183 [DOI] [PubMed] [Google Scholar]
  • 53.Cunha GR, Donjacour AA, Cooke PS, Mee S, Bigsby RM, Higgins SJ and Sugimura Y (1987) The endocrinology and developmental biology of the prostate. Endocr. Rev 8, 338–362 [DOI] [PubMed] [Google Scholar]
  • 54.Donjacour AA and Cunha GR (1993) Assessment of prostatic protein secretion in tissue recombinants made of urogenital sinus mesenchyme and urothelium from normal or androgen-insensitive mice. Endocrinology 132, 2342–2350 [DOI] [PubMed] [Google Scholar]
  • 55.Peterson RS, Clevidence DE, Ye H and Costa RH (1997) Hepatocyte nuclear factor-3α promoter regulation involves recognition by cell-specific factors, thyroid transcription factor-1, and autoactivation. Cell Growth Differ. 8, 69–82 [PubMed] [Google Scholar]
  • 56.Kopachik W, Hayward SW and Cunha GR (1998) Expression of hepatocyte nuclear factor-3α in rat prostate, seminal vesicle, and bladder. Dev. Dyn 211, 131–140 [DOI] [PubMed] [Google Scholar]
  • 57.Mirosevich J, Gao N and Matusik RJ (2005) Expression of Foxa transcription factors in the developing and adult murine prostate. Prostate 62, 339–352 [DOI] [PubMed] [Google Scholar]
  • 58.Gao N, Ishii K, Mirosevich J, Kuwajima S, Oppenheimer SR, Roberts RL, Jiang M, Yu X, Shappell SB, Caprioli RM et al. (2005) Forkhead box A1 regulates prostate ductal morphogenesis and promotes epithelial cell maturation. Development 132, 3431–3443 [DOI] [PubMed] [Google Scholar]
  • 59.Mirosevich J, Gao N, Gupta A, Shappell SB, Jove R and Matusik RJ (2006) Expression and role of Foxa proteins in prostate cancer. Prostate 66, 1013–1028 [DOI] [PubMed] [Google Scholar]
  • 60.Bhatia-Gaur R, Donjacour AA, Sciavolino PJ, Kim M, Desai N, Young P, Norton CR, Gridley T, Cardiff RD, Cunha GR et al. (1999) Roles for Nkx3.1 in prostate development and cancer. Genes Dev. 13, 966–977 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Fan L, Pepicelli CV, Dibble CC, Catbagan W, Zarycki JL, Laciak R, Gipp J, Shaw A, Lamm ML, Munoz A et al. (2004) Hedgehog signaling promotes prostate xenograft tumor growth. Endocrinology 145, 3961–3970 [DOI] [PubMed] [Google Scholar]
  • 62.Karhadkar SS, Bova GS, Abdallah N, Dhara S, Gardner D, Maitra A, Isaacs JT, Berman DM and Beachy PA (2004) Hedgehog signalling in prostate regeneration, neoplasia and metastasis. Nature 431, 707–712 [DOI] [PubMed] [Google Scholar]
  • 63.Sanchez P, Hernandez AM, Stecca B, Kahler AJ, DeGueme AM, Barrett A, Beyna M, Datta MW, Datta S and Ruiz i Altaba A (2004) Inhibition of prostate cancer proliferation by interference with SONIC HEDGEHOG-GLI1 signaling. Proc. Natl. Acad. Sci. U.S.A 101, 12561–12566 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Badve S, Turbin D, Thorat MA, Morimiya A, Nielsen TO, Perou CM, Dunn S, Huntsman DG and Nakshatri H (2007) FOXA1 expression in breast cancer-correlation with luminal subtype A and survival. Clin. Cancer Res 13, 4415–4421 [DOI] [PubMed] [Google Scholar]
  • 65.Habashy HO, Powe DG, Rakha EA, Ball G, Paish C, Gee J, Nicholson RI and Ellis IO (2008) Forkhead-box A1 (FOXA1) expression in breast cancer and its prognostic significance. Eur. J. Cancer 44, 1541–1551 [DOI] [PubMed] [Google Scholar]
  • 66.Sorlie T, Tibshirani R, Parker J, Hastie T, Marron JS, Nobel A, Deng S, Johnsen H, Pesich R, Geisler S et al. (2003) Repeated observation of breast tumor subtypes in independent gene expression data sets. Proc. Natl. Acad. Sci. U.S.A 100, 8418–8423 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Feng Y, Manka D, Wagner KU and Khan SA (2007) Estrogen receptor-a expression in the mammary epithelium is required for ductal and alveolar morphogenesis in mice. Proc. Natl. Acad. Sci. U.S.A 104, 14718–14723 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Mueller SO, Clark JA, Myers PH and Korach KS (2002) Mammary gland development in adult mice requires epithelial and stromal estrogen receptor α. Endocrinology 143, 2357–2365 [DOI] [PubMed] [Google Scholar]
  • 69.Asselin-Labat ML, Sutherland KD, Barker H, Thomas R, Shackleton M, Forrest NC, Hartley L, Robb L, Grosveld FG, van der Wees J et al. (2007) Gata-3 is an essential regulator of mammary-gland morphogenesis and luminal-cell differentiation. Nat. Cell Biol 9, 201–209 [DOI] [PubMed] [Google Scholar]
  • 70.Kouros-Mehr H, Slorach EM, Sternlicht MD and Werb Z (2006) GATA-3 maintains the differentiation of the luminal cell fate in the mammary gland. Cell 127, 1041–1055 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Usary J, Llaca V, Karaca G, Presswala S, Karaca M, He X, Langerod A, Karesen R, Oh DS, Dressler LG et al. (2004) Mutation of GATA3 in human breast tumors. Oncogene 23, 7669–7678 [DOI] [PubMed] [Google Scholar]
  • 72.Manuylov NL, Smagulova FO and Tevosian SG (2007) Fog2 excision in mice leads to premature mammary gland involution and reduced Esr1 gene expression. Oncogene 26, 5204–5213 [DOI] [PubMed] [Google Scholar]
  • 73.Bernardo GM, Lozada KL, Miedler JD, Harburg G, Hewitt SC, Mosley JD, Godwin AK, Korach KS, Visvader JE, Kaestner KH et al. (2010) FOXA1 is an essential determinant of ERα expression and mammary ductal morphogenesis. Development 137, 2045–2054 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Brisken C, Park S, Vass T, Lydon JP, O’Malley BW and Weinberg RA (1998) A paracrine role for the epithelial progesterone receptor in mammary gland development. Proc. Natl. Acad. Sci. U.S.A 95, 5076–5081 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Mallepell S, Krust A, Chambon P and Brisken C (2006) Paracrine signaling through the epithelial estrogen receptor α is required for proliferation and morphogenesis in the mammary gland. Proc. Natl. Acad. Sci. U.S.A 103, 2196–2201 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Neben K, Schnittger S, Brors B, Tews B, Kokocinski F, Haferlach T, Muller J, Hahn M, Hiddemann W, Eils R et al. (2004) Distinct gene expression patterns associated with FLT3- and NRAS-activating mutations in acute myeloid leukemia with normal karyotype. Oncogene 24, 1580–1588 [DOI] [PubMed] [Google Scholar]
  • 77.Nucera C, Eeckhoute J, Finn S, Carroll JS, Ligon AH, Priolo C, Fadda G, Toner M, Sheils O, Attard M et al. (2009) FOXA1 is a potential oncogene in anaplastic thyroid carcinoma. Clin. Cancer Res 15, 3680–3689 [DOI] [PubMed] [Google Scholar]
  • 78.Lin L, Miller CT, Contreras JI, Prescott MS, Dagenais SL, Wu R, Yee J, Orringer MB, Misek DE, Hanash SM et al. (2002) The hepatocyte nuclear factor 3α gene, HNF3α (FOXA1), on chromosome band 14q13 is amplified and overexpressed in esophageal and lung adenocarcinomas. Cancer Res. 62, 5273–5279 [PubMed] [Google Scholar]
  • 79.Sano M, Aoyagi K, Takahashi H, Kawamura T, Mabuchi T, Igaki H, Tachimori Y, Kato H, Ochiai A, Honda H et al. (2010) Forkhead box A1 transcriptional pathway in KRT7-expressing esophageal squamous cell carcinomas with extensive lymph node metastasis. Int. J. Oncol 36, 321–330 [PubMed] [Google Scholar]
  • 80.Song Y, Washington MK and Crawford HC (2010) Loss of FOXA1/2 is essential for the epithelial-to-mesenchymal transition in pancreatic cancer. Cancer Res. 70, 2115–2125 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Pasca di Magliano M and Hebrok M (2003) Hedgehog signalling in cancer formation and maintenance. Nat. Rev. Cancer 3, 903–911 [DOI] [PubMed] [Google Scholar]
  • 82.Dahmane N, Sanchez P, Gitton Y, Palma V, Sun T, Beyna M, Weiner H and Ruiz i Altaba A (2001) The Sonic Hedgehog-Gli pathway regulates dorsal brain growth and tumorigenesis. Development 128, 5201–5212 [DOI] [PubMed] [Google Scholar]
  • 83.Kalkuhl A, Kaestner K, Buchmann A and Schwarz M (1996) Expression of hepatocyte-enriched nuclear transcription factors in mouse liver tumours. Carcinogenesis 17, 609–612 [DOI] [PubMed] [Google Scholar]
  • 84.Lazarevich NL, Cheremnova OA, Varga EV, Ovchinnikov DA, Kudrjavtseva EI, Morozova OV, Fleishman DI, Engelhardt NV and Duncan SA (2004) Progression of HCC in mice is associated with a downregulation in the expression of hepatocyte nuclear factors. Hepatology 39, 1038–1047 [DOI] [PubMed] [Google Scholar]
  • 85.Coulouarn C, Factor VM, Andersen JB, Durkin ME and Thorgeirsson SS (2009) Loss of miR-122 expression in liver cancer correlates with suppression of the hepatic phenotype and gain of metastatic properties. Oncogene 28, 3526–3536 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Nitsch D, Boshart M and Schutz G (1993) Activation of the tyrosine aminotransferase gene is dependent on synergy between liver-specific and hormone-responsive elements. Proc. Natl. Acad. Sci. U.S.A 90, 5479–5483 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.O’Brien RM, Noisin EL, Suwanichkul A, Yamasaki T, Lucas PC, Wang JC, Powell DR and Granner DK (1995) Hepatic nuclear factor 3-and hormone-regulated expression of the phosphoenolpyruvate carboxykinase and insulin-like growth factor-binding protein 1 genes. Mol. Cell. Biol 15, 1747–1758 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Roux J, Pictet R and Grange T (1995) Hepatocyte nuclear factor 3 determines the amplitude of the glucocorticoid response of the rat tyrosine aminotransferase gene. DNA Cell Biol. 14, 385–396 [DOI] [PubMed] [Google Scholar]
  • 89.Wang JC, Stromstedt PE, O’Brien RM and Granner DK (1996) Hepatic nuclear factor 3 is an accessory factor required for the stimulation of phosphoenolpyruvate carboxykinase gene transcription by glucocorticoids. Mol. Endocrinol 10, 794–800 [DOI] [PubMed] [Google Scholar]
  • 90.Christoffels VM, Grange T, Kaestner KH, Cole TJ, Darlington GJ, Croniger CM and Lamers WH (1998) Glucocorticoid receptor, C/EBP HNF3, and protein kinase A coordinately activate the glucocorticoid response unit of the carbamoylphosphate synthetase I gene. Mol. Cell. Biol 18, 6305–6315 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Stafford JM, Wilkinson JC, Beechem JM and Granner DK (2001) Accessory factors facilitate the binding of glucocorticoid receptor to the phosphoenolpyruvate carboxykinase gene promoter. J. Biol. Chem 276, 39885–39891 [DOI] [PubMed] [Google Scholar]
  • 92.Schoneveld OJ, Gaemers IC, Das AT, Hoogenkamp M, Renes J, Ruijter JM and Lamers WH (2004) Structural requirements of the glucocorticoid-response unit of the carbamoyl-phosphate synthase gene. Biochem. J 382, 463–470 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Holmqvist PH, Belikov S, Zaret KS and Wrange O (2005) FoxA1 binding to the MMTV LTR modulates chromatin structure and transcription. Exp. Cell Res 304, 593–603 [DOI] [PubMed] [Google Scholar]
  • 94.Belikov S, Astrand C and Wrange O (2009) FoxA1 binding directs chromatin structure and the functional response of a glucocorticoid receptor-regulated promoter. Mol. Cell. Biol 29, 5413–5425 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Uen YH, Ko PH, Yin PH, Liu TY, Chi CW and Lui WY (2008) Glucocorticoid protects hepatoma cells against metabolic stress-induced cell death. Int. J. Oncol 33, 1263–1270 [PubMed] [Google Scholar]
  • 96.Kasper S, Sheppard PC, Yan Y, Pettigrew N, Borowsky AD, Prins GS, Dodd JG, Duckworth ML and Matusik RJ (1998) Development, progression, and androgen-dependence of prostate tumors in probasin-large T antigen transgenic mice: a model for prostate cancer. Lab. Invest 78, 319–333 [PubMed] [Google Scholar]
  • 97.Greenberg NM, DeMayo F, Finegold MJ, Medina D, Tilley WD, Aspinall JO, Cunha GR, Donjacour AA, Matusik RJ and Rosen JM (1995) Prostate cancer in a transgenic mouse. Proc. Natl. Acad. Sci. U.S.A 92, 3439–3443 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Gingrich JR, Barrios RJ, Morton RA, Boyce BF, DeMayo FJ, Finegold MJ, Angelopoulou R, Rosen JM and Greenberg NM (1996) Metastatic prostate cancer in a transgenic mouse. Cancer Res. 56, 4096–4102 [PubMed] [Google Scholar]
  • 99.Chiaverotti T, Couto SS, Donjacour A, Mao JH, Nagase H, Cardiff RD, Cunha GR and Balmain A (2008) Dissociation of epithelial and neuroendocrine carcinoma lineages in the transgenic adenocarcinoma of mouse prostate model of prostate cancer. Am. J. Pathol 172, 236–246 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Slovin SF (2006) Neuroendocrine differentiation in prostate cancer: a sheep in wolf’s clothing? Nat. Clin. Pract. Urol 3, 138–144 [DOI] [PubMed] [Google Scholar]
  • 101.van der Heul-Nieuwenhuijsen L, Dits NF and Jenster G (2009) Gene expression of forkhead transcription factors in the normal and diseased human prostate. BJU Int. 103, 1574–1580 [DOI] [PubMed] [Google Scholar]
  • 102.Jain RK, Mehta RJ, Nakshatri H, Idrees MT and Badve SS (2011) High-level expression of forkhead-box protein A1 in metastatic prostate cancer. Histopathology 58, 766–772 [DOI] [PubMed] [Google Scholar]
  • 103.Sahu B, Laakso M, Ovaska K, Mirtti T, Lundin J, Rannikko A, Sankila A, Turunen JP, Lundin M, Konsti J et al. (2011) Dual role of FoxA1 in androgen receptor binding to chromatin, androgen signalling and prostate cancer. EMBO J. 30, 3962–3976 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Robbins CM, Tembe WA, Baker A, Sinari S, Moses TY, Beckstrom-Sternberg S, Beckstrom-Sternberg J, Barrett M, Long J, Chinnaiyan A et al. (2010) Copy number and targeted mutational analysis reveals novel somatic events in metastatic prostate tumors. Genome Res. 21, 47–55 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Debes JD and Tindall DJ (2004) Mechanisms of androgen-refractory prostate cancer. N. Engl. J. Med 351, 1488–1490 [DOI] [PubMed] [Google Scholar]
  • 106.Gao N, Zhang J, Rao MA, Case TC, Mirosevich J, Wang Y, Jin R, Gupta A, Rennie PS and Matusik RJ (2003) The role of hepatocyte nuclear factor-3α (Forkhead Box A1) and androgen receptor in transcriptional regulation of prostatic genes. Mol. Endocrinol 17, 1484–1507 [DOI] [PubMed] [Google Scholar]
  • 107.Wang Q, Li W, Liu XS, Carroll JS, Janne OA, Keeton EK, Chinnaiyan AM, Pienta KJ and Brown M (2007) A hierarchical network of transcription factors governs androgen receptor-dependent prostate cancer growth. Mol. Cell 27, 380–392 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Jia L, Berman BP, Jariwala U, Yan X, Cogan JP, Walters A, Chen T, Buchanan G, Frenkel B and Coetzee GA (2008) Genomic androgen receptor-occupied regions with different functions, defined by histone acetylation, coregulators and transcriptional capacity. PLoS ONE 3, e3645. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Lee HJ, Hwang M, Chattopadhyay S, Choi HS and Lee K (2008) Hepatocyte nuclear factor-3α (HNF-3α) negatively regulates androgen receptor transactivation in prostate cancer cells. Biochem. Biophys. Res. Commun 367, 481–486 [DOI] [PubMed] [Google Scholar]
  • 110.Wang D, Garcia-Bassets I, Benner C, Li W, Su X, Zhou Y, Qiu J, Liu W, Kaikkonen MU, Ohgi KA et al. (2011) Reprogramming transcription by distinct classes of enhancers functionally defined by eRNA. Nature 474, 390–394 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Wang Q, Li W, Zhang Y, Yuan X, Xu K, Yu J, Chen Z, Beroukhim R, Wang H, Lupien M et al. (2009) Androgen receptor regulates a distinct transcription program in androgen-independent prostate cancer. Cell 138, 245–256 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.McMullin RP, Dobi A, Mutton LN, Orosz A, Maheshwari S, Shashikant CS and Bieberich CJ (2009) A FOXA1-binding enhancer regulates Hoxb13 expression in the prostate gland. Proc. Natl. Acad. Sci. U.S.A 107, 98–103 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Sreenath T, Orosz A, Fujita K and Bieberich CJ (1999) Androgen-independent expression of hoxb-13 in the mouse prostate. Prostate 41, 203–207 [DOI] [PubMed] [Google Scholar]
  • 114.Sun Q, Yu X, Degraff DJ and Matusik RJ (2009) Upstream stimulatory factor 2, a novel FoxA1-interacting protein, is involved in prostate-specific gene expression. Mol. Endocrinol 23, 2038–2047 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Jia L, Landan G, Pomerantz M, Jaschek R, Herman P, Reich D, Yan C, Khalid O, Kantoff P, Oh W et al. (2009) Functional enhancers at the gene-poor 8q24 cancer-linked locus. PLoS Genet. 5, e1000597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Lupien M, Eeckhoute J, Meyer CA, Wang Q, Zhang Y, Li W, Carroll JS, Liu XS and Brown M (2008) FoxA1 translates epigenetic signatures into enhancer-driven lineage-specific transcription. Cell 132, 958–970 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Eeckhoute J, Lupien M, Meyer CA, Verzi MP, Shivdasani RA, Liu XS and Brown M (2009) Cell-type selective chromatin remodeling defines the active subset of FOXA1-bound enhancers. Genome Res. 19, 372–380 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Yu X, Gupta A, Wang Y, Suzuki K, Mirosevich J, Orgebin-Crist MC and Matusik RJ (2005) Foxa1 and Foxa2 interact with the androgen receptor to regulate prostate and epididymal genes differentially. Ann. N. Y. Acad. Sci 1061, 77–93 [DOI] [PubMed] [Google Scholar]
  • 119.Zhang Y, Ali TZ, Zhou H, D’Souza DR, Lu Y, Jaffe J, Liu Z, Passaniti A and Hamburger AW (2010) ErbB3 binding protein 1 represses metastasis-promoting gene anterior gradient protein 2 in prostate cancer. Cancer Res. 70, 240–248 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Liu D, Rudland PS, Sibson DR, Platt-Higgins A and Barraclough R (2005) Human homologue of cement gland protein, a novel metastasis inducer associated with breast carcinomas. Cancer Res. 65, 3796–3805 [DOI] [PubMed] [Google Scholar]
  • 121.Zheng W, Rosenstiel P, Huse K, Sina C, Valentonyte R, Mah N, Zeitlmann L, Grosse J, Ruf N, Nurnberg P et al. (2006) Evaluation of AGR2 and AGR3 as candidate genes for inflammatory bowel disease. Genes Immun. 7, 11–18 [DOI] [PubMed] [Google Scholar]
  • 122.Zhang Y, Wang XW, Jelovac D, Nakanishi T, Yu MH, Akinmade D, Goloubeva O, Ross DD, Brodie A and Hamburger AW (2005) The ErbB3-binding protein Ebp1 suppresses androgen receptor-mediated gene transcription and tumorigenesis of prostate cancer cells. Proc. Natl. Acad. Sci. U.S.A 102, 9890–9895 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Scharer CD, McCabe CD, Ali-Seyed M, Berger MF, Bulyk ML and Moreno CS (2009) Genome-wide promoter analysis of the SOX4 transcriptional network in prostate cancer cells. Cancer Res. 69, 709–717 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Perou CM, Sorlie T, Eisen MB, van de Rijn M, Jeffrey SS Rees CA, Pollack JR, Ross DT, Johnsen H, Akslen LA et al. (2000) Molecular portraits of human breast tumours. Nature 406, 747–752 [DOI] [PubMed] [Google Scholar]
  • 125.Sorlie T, Perou CM, Tibshirani R, Aas T, Geisler S, Johnsen H, Hastie T, Eisen MB, van de Rijn M, Jeffrey SS et al. (2001) Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc. Natl. Acad. Sci. U.S.A. 98, 10869–10874 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Early Breast Cancer Triallists’ Collaborative Group (2005) Effects of chemotherapy and hormonal therapy for early breast cancer on recurrence and 15-year survival: an overview of the randomised trials. Lancet 365, 1687–1717 [DOI] [PubMed] [Google Scholar]
  • 127.Rakha EA, Reis-Filho JS and Ellis IO (2008) Basal-like breast cancer: a critical review. J. Clin. Oncol 26, 2568–2581 [DOI] [PubMed] [Google Scholar]
  • 128.Wolf I, Bose S, Williamson EA, Miller CW, Karlan BY and Koeffler HP (2006) FOXA1: Growth inhibitor and a favorable prognostic factor in human breast cancer. Int. J. Cancer 120, 1013–1022 [DOI] [PubMed] [Google Scholar]
  • 129.Thorat MA, Marchio C, Morimiya A, Savage K, Nakshatri H, Reis-Filho JS and Badve S (2008) Forkhead box A1 expression in breast cancer is associated with luminal subtype and good prognosis. J. Clin. Pathol 61, 327–332 [DOI] [PubMed] [Google Scholar]
  • 130.Albergaria A, Paredes J, Sousa B, Milanezi F, Carneiro V, Bastos J, Costa S, Vieira D, Lopes N, Lam EW et al. (2009) Expression of FOXA1 and GATA-3 in breast cancer: the prognostic significance in hormone receptor-negative tumours. Breast Cancer Res. 11, R40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Ademuyiwa FO, Thorat MA, Jain RK, Nakshatri H and Badve S (2009) Expression of Forkhead-box protein A1, a marker of luminal A type breast cancer, parallels low Oncotype DX 21-gene recurrence scores. Mod. Pathol 23, 270–275 [DOI] [PubMed] [Google Scholar]
  • 132.Mehta RJ, Jain RK, Leung S, Choo J, Nielsen T, Huntsman D, Nakshatri H and Badve S (2011) FOXA1 is an independent prognostic marker for ER-positive breast cancer. Breast Cancer Res. Treat, doi 10.1007/s10549-011-1482-6 [DOI] [PubMed] [Google Scholar]
  • 133.Hu X, Stern HM, Ge L, O’Brien C, Haydu L, Honchell CD, Haverty PM, Peters BA, Wu TD, Amler LC et al. (2009) Genetic alterations and oncogenic pathways associated with breast cancer subtypes. Mol. Cancer Res 7, 511–522 [DOI] [PubMed] [Google Scholar]
  • 134.Doane AS, Danso M, Lal P, Donaton M, Zhang L, Hudis C and Gerald WL (2006) An estrogen receptor-negative breast cancer subset characterized by a hormonally regulated transcriptional program and response to androgen. Oncogene 25, 3994–4008 [DOI] [PubMed] [Google Scholar]
  • 135.Hall RE, Birrell SN, Tilley WD and Sutherland RL (1994) MDA-MB-453, an androgen-responsive human breast carcinoma cell line with high level androgen receptor expression. Eur. J. Cancer 30A, 484–490 [DOI] [PubMed] [Google Scholar]
  • 136.Naderi A and Hughes-Davies L (2008) A functionally significant cross-talk between androgen receptor and ErbB2 pathways in estrogen receptor negative breast cancer. Neoplasia 10, 542–548 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Robinson JL, Macarthur S, Ross-Innes CS, Tilley WD, Neal DE, Mills IG and Carroll JS (2011) Androgen receptor driven transcription in molecular apocrine breast cancer is mediated by FoxA1. EMBO J. 30, 3019–3027 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Ni M, Chen Y, Lim E, Wimberly H, Bailey ST, Imai Y, Rimm DL, Liu XS and Brown M (2011) Targeting androgen receptor in estrogen receptor-negative breast cancer. Cancer Cell 20, 119–131 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Robyr D, Gegonne A, Wolffe AP and Wahli W (2000) Determinants of vitellogenin B1 promoter architecture. HNF3 and estrogen responsive transcription within chromatin. J. Biol. Chem 275,28291–28300 [DOI] [PubMed] [Google Scholar]
  • 140.Beck S, Sommer P, dos Santos Silva E, Blin N and Gott P (1999) Hepatocyte nuclear factor 3 (winged helix domain) activates trefoil factor gene TFF1 through a binding motif adjacent to the TATAA box. DNA Cell Biol. 18, 157–164 [DOI] [PubMed] [Google Scholar]
  • 141.Carroll JS, Liu XS, Brodsky AS, Li W, Meyer CA, Szary AJ, Eeckhoute J, Shao W, Hestermann EV, Geistlinger TR et al. (2005) Chromosome-wide mapping of estrogen receptor binding reveals long-range regulation requiring the forkhead protein FoxA1. Cell 122, 33–43 [DOI] [PubMed] [Google Scholar]
  • 142.Laganière J, Deblois G, Lefebvre C, Bataille AR, Robert F and Giguere V (2005) From the cover: location analysis of estrogen receptor α target promoters reveals that FOXA1 defines a domain of the estrogen response. Proc. Natl. Acad. Sci. U.S.A 102, 11651–11656 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Carroll JS, Meyer CA, Song J, Li W, Geistlinger TR, Eeckhoute J, Brodsky AS, Keeton EK, Fertuck KC, Hall GF et al. (2006) Genome-wide analysis of estrogen receptor binding sites. Nat. Genet 38, 1289–1297 [DOI] [PubMed] [Google Scholar]
  • 144.Lin CY, Vega VB, Thomsen JS, Zhang T, Kong SL, Xie M, Chiu KP, Lipovich L, Barnett DH, Stossi F et al. (2007) Whole-genome cartography of estrogen receptor α binding sites. PLoS Genet. 3, e87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Charn TH, Liu ET, Chang EC, Lee YK, Katzenellenbogen JA and Katzenellenbogen BS (2010) Genome-wide dynamics of chromatin binding of estrogen receptors α and β: mutual restriction and competitive site selection. Mol. Endocrinol 24, 47–59 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Hurtado A, Holmes KA, Ross-Innes CS, Schmidt D and Carroll JS (2011) FOXA1 is a key determinant of estrogen receptor function and endocrine response. Nat. Genet 43, 27–33 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Welboren WJ, van Driel MA, Janssen-Megens EM, van Heeringen SJ, Sweep FC, Span PN and Stunnenberg HG (2009) ChIP-Seq of ERα and RNA polymerase II defines genes differentially responding to ligands. EMBO J. 28, 1418–1428 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Malik S, Jiang S, Garee JP, Verdin E, Lee AV, O’Malley BW, Zhang M, Belaguli NS and Oesterreich S (2010) Histone deacetylase 7 and FoxA1 in estrogen-mediated repression of RPRM. Mol. Cell. Biol 30, 399–412 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Bretschneider N, Brand H, Miller N, Lowery AJ, Kerin MJ, Gannon F and Denger S (2008) Estrogen induces repression of the breast cancer and salivary gland expression gene in an estrogen receptor α-dependent manner. Cancer Res. 68, 106–114 [DOI] [PubMed] [Google Scholar]
  • 150.Yamaguchi N, Ito E, Azuma S, Honma R, Yanagisawa Y, Nishikawa A, Kawamura M, Imai J, Tatsuta K, Inoue J et al. (2008) FoxA1 as a lineage-specific oncogene in luminal type breast cancer. Biochem. Biophys. Res. Commun 365, 711–717 [DOI] [PubMed] [Google Scholar]
  • 151.Eeckhoute J, Carroll JS, Geistlinger TR, Torres-Arzayus MI and Brown M (2006) A cell-type-specific transcriptional network required for estrogen regulation of cyclin D1 and cell cycle progression in breast cancer. Genes Dev. 20, 2513–2526 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Gevry N, Hardy S, Jacques PE, Laflamme L, Svotelis A, Robert F and Gaudreau L (2009) Histone H2A.Z is essential for estrogen receptor signaling. Genes Dev. 23, 1522–1533 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Chan CS and Song JS (2008) CCCTC-binding factor confines the distal action of estrogen receptor. Cancer Res. 68, 9041–9049 [DOI] [PubMed] [Google Scholar]
  • 154.Zhang Y, Liang J, Li Y, Xuan C, Wang F, Wang D, Shi L, Zhang D and Shang Y (2010) CCCTC-binding factor acts upstream of FOXA1 and demarcates the genomic response to estrogen. J. Biol. Chem 285, 28604–28613 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Fullwood MJ, Liu MH, Pan YF, Liu J, Xu H, Mohamed YB, Orlov YL, Velkov S, Ho A, Mei PH et al. (2009) An oestrogen-receptor-α-bound human chromatin interactome. Nature 462, 58–64 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Frasor J, Danes JM, Komm B, Chang KC, Lyttle CR and Katzenellenbogen BS (2003) Profiling of estrogen up- and down-regulated gene expression in human breast cancer cells: insights into gene networks and pathways underlying estrogenic control of proliferation and cell phenotype. Endocrinology 144, 4562–4574 [DOI] [PubMed] [Google Scholar]
  • 157.Oh DS, Troester MA, Usary J, Hu Z, He X, Fan C, Wu J, Carey LA and Perou CM (2006) Estrogen-regulated genes predict survival in hormone receptor-positive breast cancers. J. Clin. Oncol 24, 1656–1664 [DOI] [PubMed] [Google Scholar]
  • 158.McCune K, Bhat-Nakshatri P, Thorat MA, Nephew KP, Badve S and Nakshatri H (2010) Prognosis of hormone-dependent breast cancers: implications of the presence of dysfunctional transcriptional networks activated by insulin via the immune transcription factor T-bet. Cancer Res. 70, 685–696 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Su JL, Yang CY, Zhao M, Kuo ML and Yen ML (2007) Forkhead proteins are critical for bone morphogenetic protein-2 regulation and anti-tumor activity of resveratrol. J. Biol. Chem 282, 19385–19398 [DOI] [PubMed] [Google Scholar]
  • 160.Krum SA, Miranda-Carboni GA, Lupien M, Eeckhoute J, Carroll JS and Brown M (2008) Unique ERα cistromes control cell type-specific gene regulation. Mol. Endocrinol 22, 2393–2406 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Sudo H, Kodama HA, Amagai Y, Yamamoto S and Kasai S (1983) In vitro differentiation and calcification in a new clonal osteogenic cell line derived from newborn mouse calvaria. J. Cell Biol 96, 191–198 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Nakshatri H and Badve S (2007) FOXA1 as a therapeutic target for breast cancer. Expert Opin. Ther. Targets 11, 507–514 [DOI] [PubMed] [Google Scholar]
  • 163.Fu X, Huang C and Schiff R (2011) More on FOX News: FOXA1 on the horizon of estrogen receptor function and endocrine response. Breast Cancer Res. 13, 307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Sircoulomb F, Bekhouche I, Finetti P, Adelaide J, Ben Hamida A, Bonansea J, Raynaud S, Innocenti C, Charafe-Jauffret E, Tarpin C et al. (2010) Genome profiling of ERBB2-amplified breast cancers. BMC Cancer 10, 539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Song L, Xu Z, Zhang C, Qiao X and Huang C (2009) Up-regulation of the HSP72 by Foxa1 in MCF-7 human breast cancer cell line. Biochem. Biophys. Res. Commun 386, 30–34 [DOI] [PubMed] [Google Scholar]
  • 166.Ciocca DR, Clark GM, Tandon AK, Fuqua SA, Welch WJ and McGuire WL (1993) Heat shock protein hsp70 in patients with axillary lymph node-negative breast cancer: prognostic implications. J. Natl Cancer Inst 85, 570–574 [DOI] [PubMed] [Google Scholar]
  • 167.Oloumi A, Maidan M, Lock FE, Tearle H, McKinney S, Muller WJ, Aparicio SA and Dedhar S (2010) Cooperative signaling between Wnt1 and integrin-linked kinase induces accelerated breast tumor development. Breast Cancer Res. 12, R38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Sinner D, Rankin S, Lee M and Zorn AM (2004) Sox17 and β-catenin cooperate to regulate the transcription of endodermal genes. Development 131, 3069–3080 [DOI] [PubMed] [Google Scholar]
  • 169.Berx G and Van Roy F (2001) The E-cadherin/catenin complex: an important gatekeeper in breast cancer tumorigenesis and malignant progression. Breast Cancer Res. 3, 289–293 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Chu IM, Hengst L and Slingerland JM (2008) The Cdk inhibitor p27 in human cancer: prognostic potential and relevance to anticancer therapy. Nat. Rev. Cancer 8, 253–267 [DOI] [PubMed] [Google Scholar]
  • 171.Liu YN, Lee WW, Wang CY, Chao TH, Chen Y and Chen JH (2005) Regulatory mechanisms controlling human E-cadherin gene expression. Oncogene 24, 8277–8290 [DOI] [PubMed] [Google Scholar]
  • 172.Williamson EA, Wolf I, O’Kelly J, Bose S, Tanosaki S and Koeffler HP (2006) BRCA1 and FOXA1 proteins coregulate the expression of the cell cycle-dependent kinase inhibitor p27(Kip1). Oncogene 25, 1391–1399 [DOI] [PubMed] [Google Scholar]
  • 173.Motallebipour M, Ameur A, Reddy Bysani MS, Patra K, Wallerman O, Mangion J, Barker MA, McKernan KJ, Komorowski J and Wadelius C (2009) Differential binding and co-binding pattern of FOXA1 and FOXA3 and their relation to H3K4me3 in HepG2 cells revealed by ChIP-seq. Genome Biol. 10, R129. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES